Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Fish and Shellfish Immunology 130 (2022) 294–308

Contents lists available at ScienceDirect

Fish and Shellfish Immunology


journal homepage: www.elsevier.com/locate/fsi

Bioactive immunostimulants as health-promoting feed additives in


aquaculture: A review
Seerengaraj Vijayaram a, b, Yun-Zhang Sun a, c, **, Antonio Zuorro d, Hamed Ghafarifarsani e,
Hien Van Doan f, g, *, Seyed Hossein Hoseinifar h
a
Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, China
b
Department of Environmental Studies, School of Energy Environment and Natural Resources, Madurai Kamaraj University, Madurai, India
c
The Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture, Fisheries College, Jimei University, Xiamen, China
d
Department of Chemical Engineering, Materials and Environment, Sapienza University, Via Eudossiana 18, 00184, Rome, Italy
e
Department of Fisheries, Faculty of Natural Resources, Urmia University, Urmia, Iran
f
Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai, Thailand
g
Science and Technology Research Institute, Chiang Mai University, Suthep, Muang, Chiang Mai, Thailand
h
Department of Fisheries, Faculty of Fisheries and Environmental Sciences, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran

A R T I C L E I N F O A B S T R A C T

Keywords: Bioactive immunostimulants could be derived from different sources like plants, animals, microbes, algae, yeast,
Bioactive immunostimulants etc. Bioactive immunostimulants are the most significant role to enhance aquatic production, as well as the cost
Immune system of this method, which is effective, non-toxic, and environment-friendly. These immunostimulants are supportive
Disease resistance
to increase the immune system, growth, antioxidant, anti-inflammatory, and disease resistance of aquatic ani­
Feed additives
Aquaculture
mals’ health and also improve aquatic animal feed. Diseases are mainly targeted to the immune system of aquatic
organisms in such a way that different processes of bioactive immunostimulants progress are considered
imperative techniques for the development of aquaculture production. Communicable infections are the main
problem for aquaculture, while the mortality and morbidity connected with some outbreaks significantly limit
the productivity of some sectors. Aquaculture is considered the mainly developing food production sector
globally. Protein insists is an important issue in human nutrition. Aquaculture has been an exercise for thousands
of years, and it has now surpassed capture fisheries as the most vital source of seafood in the world. Limited study
reports are available to focal point on bioactive immunostimulants in aquaculture applications. This review
report provides information on the nutritional administration of bioactive immunostimulants, their types,
functions, and beneficial impacts on aquatic animals’ health as well as for the feed quality development in the
aquaculture industry. The scope of this review combined to afford various kinds of natural derived bioactive
molecules utilization and their beneficial effects in aquaculture applications.

1. Introduction immunostimulants namely FOS, butyric acid, propionic acid, poly­


phenol, chitosan, soybean isoflavones, lentinan, lipoteichoic acid, lac­
Bioactive immunostimulants have been utilized to improve aquatic toferrin, and fucoidan have several positive effects such as immune
animal health, a broad spectrum of bioactive active molecules are used system enhancement, growth, survival and disease confrontation
to enhance aquatic animals’ health in many ways such as antimicrobial, against harmful microbes in aquatic organisms, natural bioactive
antioxidant, immunostimulant, growth-promoting, and anti- immunostimulant dietary additive technique is the new way to enhance
inflammatory properties as well as bioactive molecules applied in aquatic productions [4–8]. Microbial disease (bacteria, fungi, and vi­
aquatic feed is the novel technique, this bioactive immunostimulant ruses) outbreaks are considered the most imperative problem of aqua­
approach reliable and reproducing methods to enhance the feed quality culture production, which can eventually lead to devastating financial
[1–3]. In addition, dietary administration of various bioactive losses [11]. The modes of action of immunostimulants are boosting the

* Corresponding author. Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai, Thailand.
** Corresponding author. Xiamen Key Laboratory for Feed QualityTesting and Safety Evaluation, Fisheries College, Jimei University, Xiamen, China.
E-mail addresses: jmusunyunzhang@163.com (Y.-Z. Sun), hien.d@cmu.ac.th (H. Van Doan).

https://doi.org/10.1016/j.fsi.2022.09.011
Received 6 June 2022; Received in revised form 3 September 2022; Accepted 5 September 2022
Available online 10 September 2022
1050-4648/© 2022 Elsevier Ltd. All rights reserved.
S. Vijayaram et al. Fish and Shellfish Immunology 130 (2022) 294–308

immune system of aquatic animals and also improving the immunity (derived from aromatic plants) are widely used to enhance animal
level against harmful microbes, bioactive immunostimulants are already nutrition since these bioactive immunostimulants are applied in live­
been used to control pathogens in shrimp and fish including bacteria stock nutrition as well as in animal feed. Essential oils are an important
(Aeromonas hydrophila, Aeromonas salmonicida, Vibrio anguillarum, Vibrio bioactive immunostimulant group; as such molecules are potential
vulnificus, Vibrio salmonicida, Streptococcus sp., Yersinia ruckeri) and vi­ chemotherapeutic alternatives without side effects like immunostimu­
ruses, which cause diseases such as hematopoietic necrosis, yellow head lant, antimicrobial, anti-stress, antioxidant, and growth-stimulating ef­
virus, viral hemorrhagic septicemia and the parasite Ichthyophthirius mul­ fects while there is no ecological or harmful side effects linkage.
tifiliis, these kinds of immunostimulants are supportive to boost survival Additionally, essential oils may exhibit a positive impact on the gut
and reduced mortality rate in aquatic animals [9,10]. Aquaculture is a health of livestock as well as fish species [22]. Additionally, the colo­
vital and rapidly developing sector as it performs a very important re­ nization of gut microbiota has an important function in the regulation of
sponsibility to attain widespread protein foodstuff demands. The re­ non-specific and specific pathways of the immune system [24]. The in­
sponsibility of aquaculture to expand the socio-economic implication of testinal microbiota of fish has an extremely imperative responsibility
some places is mainly obvious since it’s not only able to provide essential due to gastrointestinal growth, higher digestive enzyme action, mucosal
nutrients but also a variety of employment opportunities are created tolerance maintenance, immunoregulation, and infection treatment
[12]. This review provides information about dietary administration, enhancement [25–28]. Similarly, the gut microbial composition changes
functions, and doses of different bioactive immunostimulants to increase in the host’s microbiota through the utilization of bioactive immunos­
healthy aquatic production and also enhance feed quality levels in the timulants from various sources [23]. Different types of dietary func­
aquaculture industry. tional feed additives such as probiotics, prebiotics, phytogenics, and
immunostimulants are used to enhance healthy aquatic animals’ pro­
2. The roles of bioactive immunostimulants in aquaculture duction and also enhance the feed quality in aquaculture, the dietary
immunogen additive increased growth performance, feed utilization,
2.1. Bioactive immunostimulants improve growth and digestive protein efficiency, and also decrease feed conversion ratio consequently,
dietary immunogen additive increased higher body protein content
Bioactive immunostimulants are present in plants, animals, and [19].
microbes or can be synthetically produced; the natural derived bioactive
immunostimulants have various biological actions such as antioxidant,
antitumor, cytotoxic, an inducer of apoptosis and cell cycle arrest, 2.3. Bioactive immunostimulants improve immunity
antimicrobial, larvicidal, chemotactic, antifouling, antimelanogenic,
and anti-inflammatory activities [13]. Bioactive immunostimulants can The nutritional additives of β-glucan, chitosan, or raffinose have
be classified into two types such as macromolecules (carbohydrates, been used to improve the non-specific immune system of koi (Cyprinus
proteins, nucleic acids, lipids, and polysaccharides) and micro molecules rubrofuscus) by amplifying various resistant criteria such as phagocytic
(natural products, primary and secondary metabolites), both the bio­ activity, respiratory burst activity, lysozyme action, WBC, and SOD ac­
molecules play a major role in aquaculture. Bioactive immunostimulants tion, finally, the bioactive immunostimulants are supportive to get
are complex organic molecules, which are present in all living organ­ better the immune system of aquatic animals [31]. MyD88 signaling
isms, while such bioactive immunostimulant substances maintain the pathway is directly triggered by MAMPs of intestinal microbiota strains.
metabolic process of all living organisms [15]. Moreover, bioactive Different kinds of substances (lysozyme, protease, complements, lectins,
immunostimulants are shaped in the body and maintain the structure and immunoglobulins) are significant for skin mucosal immunity and
and growth, while they could be characterized as necessary for body also for disease resistance in fish [29]. For example, the MyD88
functions [16]. The bioactive immunostimulants contain numerous signaling pathway plays a major part in the innate immune system
positive responses such as the immune system strengthening, antioxi­ control, while on other hand some microbial components controlled the
dant activity, mediation of hormones, and butyric acid production in the adaptive immune system, increased digestive enzyme, protein content,
colon as well as combination and/or dilution of structure in the gut, the and reduced lipid content level, there are no differences in immune gene
small intestine is not only the fatal position for nutrient digestion and expression those all the parameters confirmed by the control groups
absorption but also strongly connected with a different region of intes­ (IgM and IgT) of rainbow trout (Oncorhynchus mykiss) head kidney [30].
tinal antigens and bacteria to control gut and whole-body health [14]. Fishes treated with bioactive immunostimulants have generally
Similarly, Ashraf et al. (2020) defined bioactive immunostimulant confirmed by phagocytic cell performance increase [32], as according to
substances as the material that can cause stimulating effects on living literature the phagocytic cell action can be better by bioactive immu­
tissue [17]. nostimulants due to the activation of a natural immune response, for
example, the inflammatory reaction before adaptive response such as
2.2. Bioactive immunostimulants modulate gut microbiota antibody production, excessive doses level of some immunostimulants
provoke immunosuppression in fish. The side effects of immunostimu­
Dietary immunostimulants feed additives were described as of quite lants have not been well-studied. The study concluded that immunos­
chemical characteristic nature, containing nutritional and non- timulants are a vital role in to control of fish infections and the optimum
nutritional possessions as well as bioactive immunostimulants, which dietary immunostimulants are helpful to improve the growth of farming
may connect with direct or indirect physiological responses of fish fish culture [33]. Mendoza Rodriguez et al. (2017) examined that di­
species [18]. Moreover, dietary feed additives of carvacrol, and/or etary additives of organic acids (polyhydroxy butyrate (PHB 2%), po­
thymol koi carp (Cyprinus carpio) [20] from (Allium sativum) garlic tassium diformate (KDF 0.6%)), other biomolecules (carrageenan
rainbow trout (Oncorhynchus mykiss) [21] have revealed noteworthy (0.5%) and alginic acid (1%)) enhanced the immunological system of
possessions on gut health, digestive enzyme activity, intestinal immu­ red drum (S.ocellatus), as well as feed efficiency and weight gain, was
nity, and extra supportive photogenic targets for the progress of aquatic notably decreased in fish feed compared to the basal diet and together
feed, the dietary immunostimulant additive stimulates and modifies the bioactive immunostimulants [34]. Consequently, the bioactive immu­
intestinal microbiota conferring health benefits of the host, but notably nostimulant supplements have been shown to noticeably resilient
down-regulated the relative manner of tumor necrosis factor α (TNF-α) structure stimulation of fish [35]. In aquaculture, bioactive immunos­
(P = 0.0002) and transforming enlargement factor β (TGF-β) and timulants and their derivatives enhanced non-specific immune re­
remarkably decreased malonaldehyde (MDA) levels in fish species. sponses against various diseases, while these molecules have exhibited
Heterogeneous groups of functional feed additives, such as essential oils effective immunostimulant properties in fish and shrimps [10].

295
S. Vijayaram et al. Fish and Shellfish Immunology 130 (2022) 294–308

2.4. Bioactive compoundsas immunostimulants in aquaculture activity, and hematological and non-specific immune parameters in
beluga under experimental conditions. Finally, the optimum level of
2.4.1. Plants derivedbioactive immunostimulants 0.1% dietary polyphenol was supportive to improve fish health [51].
Plant-based bioactive immunostimulants as supplements could show
the potential impact in aquatic organisms like enhanced gut health, 2.6. Flavonoids
immune system, antimicrobial action and act as appetite stimulators, the
herbal bioactive immunostimulants only give positive effects; they Flavonoids are the most important ingredients of plants [54]. Dietary
cannot be suggested owing to their remaining and other side effects in Allium mongolicum Regel flavonoids (AMRF) supplement (40 mg/kg)
aquatic animals [36]. Similarly, the active dietary supplementation of enhanced the immune system, antioxidant status, regulation of gene
herb and spices were capable to provoke and release intestinal enzymes, activation, and infection resistance of Cephalopholis argus (Peacock hind)
mucosal serum immunities, and antioxidant response that will stimulate against Aeromonas hydrophila as well as considerably decreased in serum
the appetite; thus, rising food consumption and subsequently improving cortisol, liver MDA content, serum ALT and AST activity [55]. Dietary
the efficiency in Nile tilapia (Oreochromis niloticus), this dietary additive supplementation of R. verniciflua extract flavonoids enhanced feed
has no significant changes in feed conversion ratio and also dietary in­ quality level and fish production, exhibiting also a significant impact on
clusion was not affected to fish health [37]. In addition, various plants antiviral effects in fish viral diseases like infectious hematopoietic necrosis
derived bioactive elements such as flavonoids, alkaloids, phenolics, virus (IHNV) and viral hemorrhagic septicemia virus (VHSV) [56].
terpenoids, tannins, steroids, glycosides, saponins, and essential oils
have been described to show positive effects namely immunostimula­ 2.7. Alkaloids
tion, growth promotion, anti-pathogenic, appetite stimulation, and
anti-stress in fish species, this study pointed out that dietary additives of Alkaloids are found in plant tissues as a type of water-soluble salts of
herbal bioactive immunostimulant as helpful to enhanced physiological organic acids, esters, or sugars rather than as free bases [57]. Moreover,
and hematological responses in fishes [36]. The plant-based bioactive alkaloids contain different types of protein bands, which have a signif­
immunostimulants were commonly tonics for the release of digestive icant role in immune system enhancement and infection resistance
enzymes and have a direct effect on intestinal microflora, while ac­ against various pathogens in fish [58]. Subsequently, dietary additives
cording to literature these compounds can increase the digestibility, feed of Gelsemium elegans alkaloids (40 mg/kg) increased intestinal villus
conversion, and protein synthesis of the rainbow trout (Oncorhynchus length, villus number, muscle growth, antioxidant activity, intestinal
mykiss) [38]. Different types of plant bioactive immunostimulants have immune activity, cytokine-related genes expression, and also to
been added to feed supplements, demonstrating significant impacts in improved the disease resistance of Wuchang bream (Megalobrama
farmed animals, while such dietary additives are safe for animals, con­ amblycephala), while firmicutes abundance was reduced at phylum level
sumers, and the environment [39]. Abha and Abhisweta (2020) reported after the alkaloid feeding [59]. Zhou et al. (2016) reported that dietary
that the animal feed industry utilized plant bioactive immunostimulants additives (25 g/kg) of the fibrous root of Rhizoma Coptidis containing
to develop animal nutrition and health [40]. Additionally, Baleta et al. alkaloids could enhance the immune system and disease resistance in
(2022) suggested that dietary additives of 1% ethanolic katuk extract common carp (Cyprinus carpio) [60].
bioactive immunostimulant enhanced growth, appetite, and improved
food utilization in Orange-spotted grouper (Epinephelus coioides) [41]. 2.8. Fructooligosaccharides

2.5. Polyphenols FOS is derived from blue agave plants, vegetables, fruits, cereals, and
grains [61], composed of D-fructans, in which fructose is attached by
The polyphenols group is one of the major classifications of bioactive b-(2–1) glycosidic connections and associated with an end glucose unit.
immunostimulants produced by plants to defend against different Additionally, FOS may be linked via tolls like receptors (TLR2) pointing
pathogenic bacteria, fungi, and viruses and also defend response to to macrophages [62] and stimulating the expression of antimicrobial
various types of abiotic stress [42]. Therefore, polyphenols are sug­ peptides (Leap), which have a significant role in natural immune pro­
gested as alternative options for the (partial) replacement of chemicals tection and disease resistance in fish (Megalobrama amblycephala) [63].
and antibiotics in aquaculture development [43,44]. In addition, poly­ Likewise, dietary supplementation of FOS (1%,2%, and 3%) increased
phenol dietary additive (tannins from 2.5 to 20.0 g/kg) was used for lysozyme action, salinity stress resistance, leukocyte production, and
immunological parameters enhancement in rohu (Labeo rohita) finger­ elevated digestive enzyme function in Caspian roach fry (Rutilus rutilus)
lings, while these supplements have shown numerous positive effects [64]. Similarly, our research group described that diet containing (5
such as resistance against oxidative stress improvement, reproductive g/kg) FOS improved intestinal enzyme activity, and lysozyme action,
performance, growth performance and infection resistance in different and also decreased triglyceride (TG) levels compared to the control diet
fish species such as European sea bass (Dicentrarchus labrax), black carp in Japanese flounder (Paralichthys olivaceus) [59,65]. Dietary supple­
(Mylopharyngodon piceus), Wuchang bream (Megalobrama amblyce­ mentation of FOS (0.3 and 0.6%) has shown a beneficial effect on the
phala), common carp (Cyprinus carpio), convict cichlid (Amatitlania innate immune response of Nile tilapia (Oreochromis niloticus),
nigrofasciata) and beluga sturgeon (Huso huso) [10,45–51]. Dietary confirmed by the major progress of serum lysozyme action and IgM level
polyphenol supplement (0.2%) was extracted from chestnut and olive and also both plasma and phenoloxidase performance were noticeably
mill wastewater, this polyphenol additive improved the growth perfor­ (P < 0.05) affected only by dietary FOS levels with the maximum
mance, immunological parameters, and antioxidant status, and also feed standards experiential in fish fed 0.6 and 0.3% FOS, respectively [66].
conversion ratio of fish-fed polyphenols notably diminished and differ­
entiated from the control groups in common carp (Cyprinus carpio) fin­ 2.9. Galactooligosaccharides
gerlings [52]. Dietary Clarius lazera phenol additives have been used in
various concentrations (35, 75, 150, and 300 mg/L) and enhanced he­ Galactooligosaccharides (GOS) incorporate 2–20 different prebiotic
matological parameters and also elevated LDH, glucose, and cortisol, substances of galactose and glucose and are generally formed by the
and significantly reduced serum cholesterol concentration in catfish enzymatic hydrolysis of lactose [67]. GOS has a small point of poly­
(Siluriformes). The dietary polyphenol was supportive to improve the merization, which improves its metabolization rate by probiotics [68].
fish’s immune system [53]. In addition, dietary tannins polyphenol The use of GOS (1% and 2%) dietary supplement enhanced humoral and
additive (0.1%) was derived from chestnut wood and were in various mucosal immune response, survival rate, and increased salinity resis­
analyzes such as growth parameters, growth gene expression, enzymatic tance in Caspian roach (Rutilus rutilus) fry as well as there is no negative

296
S. Vijayaram et al. Fish and Shellfish Immunology 130 (2022) 294–308

impacts in the experimental condition [69]. Nutritional management of Geniposide showed a variety of pharmacological responses both in vitro
GOS as an immunostimulant in rockfish (Sebastes schlegeli) presented and in vivo applications, such as antidiabetic, anti-inflammatory, neu­
that feeding rockfish with (1%) of GOS for 8 weeks, notably improved roprotective, hepatoprotective, analgesic, antidepressant-like, car­
skin mucus as well as humoral immune criteria, and also this bioactive dioprotective, antioxidant, immune-regulatory, antithrombotic, and
immunostimulant not affected to the physiological and hematological antitumor activity. Geniposide is derived from Gardenia jasminoides Ellis,
responses in fish species [70]. Yukgehnaish et al. (2020) reported that a Chinese herbal medicine [85]. Nevertheless, few studies demonstrate
the dietary supplement of GOS (10 g/kg− 1) acted as a significantly the efficacy of geniposide as an immunostimulant in fish species, dietary
important prebiotic for the Atlantic salmon (Salmo salar) and also this administration of geniposide 400–800 mg/kg increased total collagen
bioactive immunostimulant affected protein concentration in the wet and alkaline-insoluble collagen content in grass carp (Ctenopharyngodon
body and protein retention were reduced by 6 and 9%, respectively, Idella) muscle than control and also fish fed diet containing geniposide
relative to the basal fish [71], Caspian roach (Rutilus rutilus) [72], notably reduced muscle crude lipid content compared to control groups
rainbow trout (Oncorhynchus mykiss) [73], common carp (Cyprinus car­ [86]. Geniposide was shown as an efficient diet supplement to control
pio) [25,74], Caspian white fish (Rutilusfrisii kutum) [75], narrow-clawed the apoptosis-related factor, reduce WSSV (white spot syndrome virus)
crayfish (Astacusleptodactylus Leptodactylus) [76], and rockfish (Sebastes simulation and increase the survival rate of WSSV-challenged red
schlegelii) [70]. swamp crayfish (Procambarus clarkii) [87]. Similar beneficial effects of
lysozyme reaction were also observed in crucian carp (Carassius car­
2.10. Inulin assius) [88], and Nile tilapia (Oreochromis niloticus) [66] feeding
geniposide-containing food. According to the literature, the use of 100
Inulin exists in several fruits, vegetables, and cereals, like leeks, mg geniposide/kg was proved positive effects on immune reaction
onions, chicory, bananas, wheat, garlic, and artichokes. It is reported improvement and infection resistance in crucian carp (Carassius car­
that dietary inulin supplement (5 g/kg) could improve hematocrit and assius), one of the most (freshwater) cultivated fish species in China.
NBT activity levels and lysozyme activity as well as there is no negative Furthermore, this dietary supplement was tested for innate immune
impact on physiological and hematological response in Nile tilapia response improvement, upregulated resistant connected gene exhibi­
(Oreochromis niloticus) [77]. Gupta et al. (2020) stated that dietary inulin tion, and infection resistance against Aeromonas hydrophila in crucian
additives can control the hematological and/or serum biochemical pa­ carp (Carassius carassius) [89].
rameters (erythrocyte counts, mean corpuscular volume (MCV), he­
matocrit, mean corpuscular hemoglobin (MCH), hemoglobin, mean 2.13. Lectins
corpuscular hemoglobin concentration (MCHC), leukocyte counts,
serum glucose or serum total protein levels, differential leukocyte counts Lectins are carbohydrate-binding proteins, present in most plants
and serum cholesterol); thus this indicator has a prominent role in fish and some animals [90]. Lectins are considered as a carbohydrate
health [78]. Furthermore, Mo et al. (2015) presented that dietary inulin detection domain, mainly binding sugars while there are different
additive (2%) improved growth action and the natural immune system classifications of animal lectins namely C-type, I-type, F-type, L-type,
of grass carp (Ctenopharyngodon Idella) [79]. Another study about inulin P-type, M-type, R-type, F-box lectins, chitinase-like lectins, galectins,
supplement (15–20 g/kg) in Barramundi (L.calcarifer) juveniles; re­ intelectins, calnexin, and ficolins. Lectins play a major role in intracel­
ported significant effects in histology, biochemical parameters, and lular and extracellular, extracellular lectins are vital for cell signaling
immunohistology [80]. and pathogen recognition while intracellular lectins play a major part in
the convey of proteins movement throughout the cells and in protein
2.11. Salidroside sorting [91]. C- type lectins, which are responsible for stimulating the
immune system, are particularly linked with PAMPs on the surfaces of
Salidroside is a glucoside of tyrosol, which is present in the plant several harmful microbes, providing them the ability to detect a broad
species of Rhodiola Rosea. It has been considered, along with rosavin, as diversity of pathogens [92]. Several classifications of lectins have been
one of the most significant factors responsible for the known anti- recognized in rainbow trout serum through calcium ion chains to
depressant and anxiolytic actions of this plant [81]. Zhang et al. A. salmonicida lipopolysaccharide [93]. On the other hand, opsonizing
(2021) reported that salidroside was found not only in the plant Rhodiola lectins, which are present in fish serum, may be beneficial for fish health,
Rosea but also in a few Chinese herbs like Angelica root [82], Ganoderma as are considered for the immune mechanism and prevention of aquatic
lucidum [83], Prunella vulgaris, etc., as well as this bioactive immunos­ animal diseases. The components play an imperative function in pro­
timulant enhanced the immune system and antiviral effects challenging tecting the host by identifying pathogens and then increasing their
against coxsackievirus B3 and dengue virus [84]. Xie et al. (2020) pointed elimination and appearance in phagocytic cells of the immune system.
out that salidroside (0.1% and1.0%) components were highly present in The mammalian mannose-associated lectin can opsonize in different
Prunella vulgaris extracts, and these factors enhanced non-specific im­ types of organisms such as yeasts, bacteria, and surrounded viruses, like
mune activity, respiratory burst activity, phagocytic activity reduced HIV or influenza [94]. Similarly, the potential function of lectins as
mortality rate, and treat Olive flounder (Paralichthysolivaceus) challenge non-recognized substances in all living organisms’ immunity [95].
with Uronema marinum. Finally [4], Yang et al. (2020) mentioned that Likewise, the crucial role of lectins in molecular biology and immuno­
dietary additive of salidroside improved the immune system in crucian logical analysis in animals [96], while the importance of lectins in the
carp (Carassius carassius) and enhanced the defense action against innate immune reaction of mammals is well known [97]. In addition,
harmful microbes, main at the concentration of 100 mg/kg for four Liang et al. (2020) pointed out that lectins from immune cells of marine
weeks. The protective effect of salidroside on crucian carp (Carassius invertebrates were effective in improving disease resistance against
auratus) might be considered as an alternative to antibiotics for con­ V. vulnificus and V. pelagicus in aquatic animals and also reduced mor­
trolling fish infections in aquaculture as well as the quantity of tality rate in aquatic animals [98].
A. hydrophila in the kidney and spleen was considerably decreased in
salidroside additive diet groups (P < 0.05). 2.14. Carotenoids

2.12. Geniposide Carotenoids are natural pigments, with a significant role in all living
organisms like plants, algae, fungi, bacteria, and animals. Animals
Geniposide is a bioactive iridoid glycoside; it is present in different cannot synthesize carotenoids, so they obtain them from their diet [99].
types of medicinal herbs, namely Gardenia jasminoides (fruits). Shrimp-extracted carotenoid supplements of 100 and 200 mg/kg could

297
S. Vijayaram et al. Fish and Shellfish Immunology 130 (2022) 294–308

lead to immune system enhancement and infection resistance of Carp leucocytes, cytokine, and macrophages [110]. In aquaculture, dietary
against A. hydrophila [100]. The addition of carotenoids in feed chitosan improved the protective effect against bacterial infections and
exhibited a beneficial role in both fish and fishery products as patho­ enhanced the immune response in hybrid striped bass (Morone saxatilis)
genic organisms’ inhibition was achieved. Carotenoids’ dietary addi­ [111], while recently, chitosan was presented as an effective immu­
tives range (50 and 100 mg/kg) varies from species to species [101]. nostimulant in different fish species [112]. Dietary supplement of chi­
Many researchers exhibited that a dietary additive of 100 mg car­ tosan enhanced non-specific immune response under experimental
otenoids/kg can boost the immune system, survival, and growth as well conditions of healthy control, and additionally, chitosan was tested
as avoid harmful effects of lipid peroxidation in different species such as against the immune response modulation in rohu (Labeo rohita) [113].
rainbow trout (Oncorhynchus mykiss), giant tiger prawn (Penaeus mon­ Diets with chitosan for rainbow trout (Oncorhynchus mykiss) [114], olive
odon) and European bullhead (Cottus gobio) [102–104]. Natural immu­ flounder (Paralichthys olivaceus) [115], koi (Cyprinus rubrofuscus) [116],
nostimulants, namely dietary supplements of carotenoids boosted the and kelp grouper (Epinephelus bruneus) [107] demonstrated that such a
immune system, disease resistance, and antioxidant function of aquatic bioactive immunostimulant could improve growth, natural immunity,
animals and also downregulated in superoxide dismutase but high infection, stress resistance, enhance immunological criterion and in­
catalase activity compared to the control one [105]. The function of crease water quality. The mechanism of chitosan action as an immu­
plants based bioactive compounds as immunostimulants in aquatic an­ nostimulant in fish has not been fully established yet. Similar studies
imals has been summarised in Table 1. showed that chitosan additives might increase the innate immune sys­
tem response of various fish species [117]. Oushani et al. (2020) re­
2.15. Animal originated bioactive immunostimulants ported the action of chitosan as an immunostimulant in rainbow trout
(Oncorhynchus mykiss), as they observed that a chitosan (2.5 g/kg) diet
2.15.1. Chitosan was favorable on immunological criteria and increased the survival rate
Chitosan is a type of alkaline polysaccharide existing in the shell of of rainbow trout (O. mykiss) against environmental stress [114]. Wu
aquatic organisms namely crabs, shrimps, and shellfish. Chitosan is a et al. (2020) reported that the immunostimulatory feature of chitosan
conventional non-toxic, safe, bio-compatible, and bio-degradable poly­ for olive flounder (Paralichthys olivaceus) activates the innate immune
mer substance. The main effect of chitosan molecule progression is to system of fish species, thus this action exhibits a defense role in the fish
significantly increase the growth of aquatic species [106], reduce the body through amino moieties [115].
growth of harmful microbes [107], purify the aquaculture water [108],
and improve the resistance of aquatic organisms to infections [106]. 2.16. Lactoferrin
Chitosan can chelate metal ions via the role of amino acid groups and
hydroxyl substances and might decrease the heavy metal ions in water; Lactoferrin is a globular glycoprotein with a molecular mass of about
thus chitosan is considered a major defender factor for the aquatic 80 kDa that is widely represented in different secretory fluids namely
species [109]. In addition, immunostimulants are responsible for the milk, saliva, tears, and nasal secretions. Lactoferrin is one of the sig­
improvement of inflammatory cell function namely polymorphonuclear nificant bioactive mechanisms of the immune system in the body; it has

Table 1
Plants originated bioactive compounds as immunostimulants in aquaculture.
Origin Bioactive Aquatic organisms Immune response References
immunostimulants

Plant Fructooligosaccharides Caspian roach ↑lysozyme function, improvement of IgM level, No significant different ACP and [63,65]
increases PO
Common carp
Nile tilapia
Japanese sea bass ↑growth, digestion and immune activities [190]
Tambaqui Promote growth and improved healthiness [191]
Juvenile grouper Epinephelus Improved immune system and intestinal morphology [192]
coioides
Galactooligosaccharides Caspian roach ↑ Humoral and mucosal immune reaction [69,71,72,
Rainbow trout 75]
Narrow-clawed
Crayfish
Common Carp Controlled innate immune response in skin mucosa [193]
Pacific white shrimp Modulate gut microbiota composition, reduce stress and enhanced immune system [194]
Inulin L. calcarifer ↑ hematocrit, NBT and lysozyme activity, biochemical parameters and immuno- [63,79]
Rainbow trout haematology
common carp ↑growth performance, innate immune response and antioxidant activity [195]
Nile tilapia Improved fish health under salinity stress condition [196]
Salidroside Crucian carp ↑ immune related gene and non specific immune system [4,105]
Paralichthysolivaceus
Geniposide Crucian carp ↑ innate immune response, upregulated immune gene expression, RPA [65,188]
Nile tilapia
Crayfish Anti inflammatory, antioxidant and antiviral activity [197]
Polyphenols Common carp fingerlings ↑haematological parameters(erythrocytes and haemoglobin), anti oxidant status [51]
Amatitlania nigrofasciata Improved humoral and mucosal response and antioxidant activity [198]
Lates Calcarifer Improved growth, mucosal immune response and liver oxidative status [199]
Flavonoids C.argus ↑immune system, antioxidant activity, regulation of immune gene expression [7,54]
Trachinotusovatus
Ctenopharyngodon idella ↑Antioxidant, anti inflammatory and immune response [200]
Alkaloids Megalobramaamblycephala ↑antioxidant activity, Intestinal immune activity, RPA [58,59]
common carp
Pacific white shrimp Improved growth response, survival rate, immune response and antibacterial [201]
activity

↑- Enhance or Increase or Improve.

298
S. Vijayaram et al. Fish and Shellfish Immunology 130 (2022) 294–308

antimicrobial activity (bacteriocide, fungicide) and is part of the innate osmotic pressure of the cytoplasm. Peptidoglycan was also involved in
defense, mainly against mucose. Lactoferrin connects with DNA and the binary division during bacterial cell reproduction [123]. Oral dietary
RNA, polysaccharides, and heparin, and performs the biological supplements of peptidoglycan (0.2 mg/kg) extracted from Bifidobacte­
response in complexes with these ligands [118]. The dietary additive of rium thermophilum could act against vibriosis and white spot syndrome in
100 mg/kg Lf enhanced the immune response, respiratory burst, leu­ kuruma shrimps (Marsupenaeus japonicas) as well as reduce mortality
kocytes peroxidase content, and phagocytic index in gilthead sea bream and increase the survival rate in shrimp [124]. Likewise, dietary addi­
(Sparus aurata) [119]. [120] Morshedi et al. (2020) observed that the tives of cellular components like cell wall (CW), peptidoglycan (PG), and
dietary supplement (1200 mg/kg) of Lf could enhance low salinity lipoteichoic acid (LTA) enhanced physiological and immunological
stress, tolerance of air exposure, natural immune response, and mucus response, while notably decreased feed conversion ratio (FCR) was
secretion of juvenile orange-spotted grouper (Epinephelus coioides), as noticed in all the treatments compared with the control group in
well as dietary LF, did not affect the growth rate of this species under the orange-spotted grouper (Epinephelus coioides) [125]. [126] Kondo et al.
condition applied in this study. Similarly [35], Luna-Castro et al. (2021) (2021) demonstrated that dietary additives of A3a-PG (8 g A3α-PG/kg)
represented that the nutritional additive of Lf at a level of 100 mg/kg led to the immune system enhancement, disease resistance, and growth
increased the innate immunity and infection resistance of sea bream performance of Japanese flounder (Paralichthys olivaceus) as well as the
(Pagrus major) and Asian catfish (Siluriformes), as well as this study, highest dose explored in the previous report, (16 g A3α-PG/kg), also
described that support the possible use of Lf as an immunostimulant for failed to show any significant influences, although no obvious pathology
farmed fish species. Several Lactoferrin (Lf) products have been classi­ was observed. Another case of dietary additive of peptidoglycan (0.18
fied and evaluated, but bovine lactoferrin (BLf) has received attention g/kg for5 days/week) exhibited an increase in the immune system and
for its potential for animal protection. Owing to its immunoregulatory infection resistance of black tiger shrimp (Penaeus monodon Fabricius)
function, BLf may stimulate antiviral, antifungal, and antibacterial and there is no significant changes to the growth [127]. The immunos­
functions and subsequently may activate efficient defense against timulatory effect of microbial peptidoglycan is acknowledged from the
different microbial infections in some fish species according to the dose laboratory verification process, which can stimulate innate immune
and administration methods [121]. Additionally, dietary Lf supple­ responses, phagocytic index, and survival rate and infection resistance
mentation of 100 mg/kg for 1 week the immune system improvement of against aquatic pathogens in aquatic animals [128].
diverse fish species such as goldfish (Carassius auratus), rainbow trout
(Oncorhynchus mykiss), Atlantic salmon (Salmo salar), red sea bream 2.18. Lipopolysaccharides
(Pagrus major) and Asian catfish (Siluriformes) [122]. The function of
animals-base bioactive compounds as immunostimulants in aquatic Lipopolysaccharides (LPS) are huge molecules incorporating a lipid
animals has been summarised in Table 2. and a polysaccharide controlled of O-antigen, outer core, and inner core
connected with a covalent bond, while they are attending in the outer
2.17. Bacterium originated bioactive immunostimulants layer membrane of the Gram-negative bacteria. LPS are the components
of the gram-negative bacteria cell wall and also turn on B cell prolifer­
2.17.1. Peptidoglycan ation. LPS injected into red sea bream (Pagrus major) exhibited improved
Peptidoglycan or murein is a polymer consisting of sugars and amino macrophage phagocytic action [129]. The LPS are very effective even in
acids that forms a mesh-like peptidoglycan layer outside the plasma extremely low doses; while they can be derived from bacteria by
membrane of most bacteria, forming the cell wall. The sugar component chemical extraction methods and present better immunostimulating
consists of alternating residues of β-(1, 4) linked N-acetyl glucosamine properties during their use in fish species [130]. Ali et al. (2021) sug­
(NAG) and N-acetylmuramic acid (NAM), and attached to the NAM is a gested that LPS (1250 μg) played a significant role in rainbow trout
peptide chain of three to five amino acids. Peptidoglycan performs the (Oncorhynchus mykiss) and carp fish (Cyprinus carpio) species, as they are
structural function, strengthens the bacterial cell wall, and prevents the good immunomodulators and beneficial response in the disease resis­
tance of fish species, the lipopolysaccharides improved phagocytic ac­
tivity, total serum Ig and get better mucus lysozyme compared to
Table 2
Animal originated bioactive compounds as immunostimulants in aquaculture. untreated fishes mortality level is increased [32,131]. Abd El-Kader
et al. (2021) recommended that dietary supplement of LPS (50 and 100
Origin Bioactive Aquatic Immune response References
immunostimulants organisms
μg of LPS/fish) was injected intraperitoneally and successfully activated
innate cellular and secondary immune response as well O-specific
Animal Chitosan Rainbow [105,
↑haematological
linkage for activating the adaptive immune system and disease resis­
trout parameters, RPA 112–114]
Olive tance against A. hydrophila [132]. In contrast, oral consumption did not
flounder alter any of the aforementioned criteria. Similarly [133], Holen et al.
Koi (2021) reported that dietary additive of lipopolysaccharides was derived
kelp from Salmonella Typhimurium. The lipopolysaccharide component
grouper
enhanced better immunostimulant, phagocytic activity, and disease
Lactoferrin Asian ↑non specific [34,117,
catfish immune response, 118] resistance against Escherichia coli and A. salmonicida disease in trout
Atlantic respiratory burst, (Oncorhynchus mykiss). In addition [134], Mohammadi et al. (2020)
salmon leukocytes reported that a nutritional LPS additive of 0.1 mL was derived from
peroxidase content
Aeromonas hydrophila enhanced disease resistance against A. hydrophila
and phagocytic
index, RPA in (Oreochromis niloticus) Nile tilapia as well as for disease prevention
gilthead during unfavorable environmental conditions.
sea bream
Carotenoids Carp ↑antioxidant [98, 2.19. Lipoteichoic acid
rainbow activity, RPA. 100–102]
trout immune system
giant tiger response, Disease Dietary supplementation of lipoteichoic acid (LTA) was derived from
prawn resistance probiotics and is used to regulate immune gene expression as well as in
European ligand binding connections. Since LTA can stimulate the immune
bullhead
response in fish, it might be favorable for the control of disease in Gram-
↑- Enhance or Increase or Improve. positive bacteria such as Streptococcus iniae and Streptococcus agalactiae

299
S. Vijayaram et al. Fish and Shellfish Immunology 130 (2022) 294–308

[135]. Similarly [136], Yang et al. (2019) observed that dietary addi­ and metabolic regulatory functions [147,149]. The nutritional addi­
tives of both peptidoglycan and LTA extracted from B. pumilus SE5 can tive of lentinan (100 μg/ml) has notably increased the intestinal anti­
stimulate TLRs/MyD88 signal pathway and the condition of antibacte­ oxidant, and inflammatory capacity by enhancing the performance of
rial agents and thus form the gut microbiota in orange-spotted grouper SOD, CAT, and GSH-Px, and suppressing lipid peroxidation and also
(E. coioides). Additionally, dietary additive of LTA extracted from pro­ lentinan may be effective for the treatment of gut inflammation
biotic B. pumilus SE5 was utilized to uncontrolled three antibacterial including IBD in juvenile taimen (Hucho taimen) [148,149]. [150].
peptides genes (epinecidin-1, hepcidin-1, and β-defensin) and immune Galindo-Villegas et al. (2016) reported that lentinan can induce anti­
gene expression (TLR2, NOD2, IL-8, IgM) in the head kidney of Grouper microbial activity and stimulate the immune system through in vitro
(Epinephelus coioides) [137]. Earlier studies have revealed that LTA is the methods. Moreover, dietary additives of lentinan (100–1000 μg/ml)
main cell wall element of Gram-positive bacteria, can associate with the increased the immune system and disease resistance in Common carp
TLR-2 receptor, and triggers a cascade of responses. L. Plantarum LTA (Cyprinus carpio) by in vitro methods [151].
extensively improved the feature of TLR2 and V. anguillarum flagellin
particularly improved TLR5. It is differentiated that LTA, flagellin, and 2.23. Mannan oligosaccharides
LPS bacterial components are mainly stimulated by TLR2, TLR5, and
TLR9 receptor pathways [138,139]. [140] Gao et al. (2016) suggested Mannan oligosaccharides (MOS) are extracted from the cell wall of
that dietary LTA enhanced the natural immune system of silver pomfret yeasts and recently researchers indicated the beneficial properties of
(Pampus argenteus). Probiotic dietary additives of LTA can regulate the MOS in fish and shrimp species [152]. The possible effect of MOS (0.6%)
activation of several inflammatory factors and provoke apoptosis to a in aquaculture content is determined by the mannose receptor (endo­
lower level, as might also be determined by the physiological charac­ cytic receptor), while these receptors communicate with macrophages
terization of the bacterium. Dietary supplements of LTA an imperative and endothelial cells to be identified together with individual glyco­
function in the host cell TLR and also enhanced immune response, proteins and microbial glycan ligands [153]. MOS (0.6%) presented a
probiotic colonization, and decreased pathogenic load while sustaining related consequence on the function of lysozyme in Nile tilapia (Oreo­
a strong and active strength among the host and its gut bacteria. chromis niloticus), Japanese flounder (Paralichthysolivaceus), Rainbow
trout (Oncorhynchus mykiss), and Channel catfish (Ictalurus punctatus),
2.20. Levan while the dietary additives of MOS were not affected in fish growth and
immune system [154]. MOS as feed supplements (6 g/kg) in aquaculture
Levan is a naturally occurring fructan, the homopolymer of fructose. of fish and shellfish may demonstrate to be supportive to the host by
The molecular weight and degree of branching of levan differ depending increasing innate immunity, although more studies described that it is
on the production organism. Moreover, the molecular weights of bac­ needed to infer the appropriate level of MOS addition to diverse fish
terial levan are much higher than the plant levan due to the multiple species and also MOS dietary supplement was significantly decreased
branching [141]. Levan is the component of natural polymeric chain lipogenic enzymes response in liver and resulted in a lower lipid
linkages, which are mainly present in the microbial extracellular prod­ vacuolization and more regular-shaped morphology of hepatocytes
ucts of Bacillus subtilis, Bacillus polymyxa, Zymomonas mobilis, Acetobacter approximately the sinusoidal spaces, which could be associated to better
xylinum, and a few plant species [78]. Similarly, dietary additives (25 energy and good feed utilization because of the improvement of feed
g/kg) of microbial levan exhibited beneficiary effects in aquatic animals utilization and the decrease of feed intake found in the current experi­
like improved non-specific immune response, disease resistance, ment [155]. The dietary additive of MOS (0.4%) notably activated he­
growth, survival, and enhanced tolerance to thermal stress, and also molytic and phagocytic reactions in rainbow trout (Oncorhynchus mykiss
levan additive diets notably reduced the count of total viable aerobic Walbaum) [156], while an amount of 2 g MOS/kg for 60 days was used
bacteria and Vibrio spp. in the gut region of orange-spotted grouper to evaluate certain immune components and immuno-associated en­
(Epinephelus coioides H.) [142]. Moreover, the dietary additive of mi­ zymes in red sea bream (Pagrus major) as well as the specific immu­
crobial levan (1.25%) enhanced the immune response and thermal stress nostimulatory activity of MOS in the gastrointestinal tract of fish and
tolerance effect of rohu (L. rohita) juveniles, this optimum dietary ad­ also the performance of lysozyme, bactericidal and peroxidase in serum
ditive can confer protection against thermal stress tolerance in fish and mucus of fish fed MOS diets improved over the control [157].
species [143]. Similarly, the dietary feed supplement of MOS (0.6%–1%) was experi­
enced to increase blood protein levels in rohu (Labeo rohita) and Nile
2.21. Fungal originated bioactive immunostimulants tilapia (Oreochromis niloticus), and also dietary additive of MOS at a
0.6% and 1% dietary level stimulates growth and survival in rohu (Labeo
Higher fungi are considered important sources of natural products rohita) and Nile tilapia (Oreochromis niloticus). In difference, elevated
and have also exhibited various biological properties like anti-microbial, addition levels of immunostimulants led to an immunosuppressive effect
anti-oxidant, immunostimulation, hypoglycaemic activities, and anti- in rohu (Labeo rohita) and Nile tilapia (Oreochromis niloticus) [158].
cancer [144]. Numerous higher fungi (Pleurotus ostreatus, Lentinula
edodes, Grifola frondosa, Coriolus Versicolor, Ganoderma lucidum, and 2.24. β-Glucans
Schizophyllum commune) have bioactive polysaccharides in their fruiting
bodies; mycelium and broth culture [145]. Consequently, poly­ Glucans are primarily present in the cell walls of bacteria, fungi,
saccharides can significantly improve the growth and natural immune algae, lichens, and yeasts they obtain a helical or spiral backbone suit­
response of fish and shrimp species [146–148]. able to specific intra-factor hydrogen bonding. These β-glucans (10 mg/
kg) have been commonly expressed to control the immune function as
2.22. Lentinan well as blood neutrophils, oxidative radical production, and superoxide
anion production in stimulated macrophages of Litopenaeusvannamei
Lentinan is a biological active transformer, which is shown to be juveniles [159]. The main component of β glucans is high-quality
effective in protecting the host from oxidative damage without obvious immunostimulating factors. The abundance and nature of side chains
adverse activity and also diet additives of lentinan were used to modify strongly affect the capability of the glucans to interfere with binding to
the gastrointestinal microbiota, which would be useful to sustain the external receptors on the particular cells influencing the reaction of the
epithelial barrier role and enhance intestinal immunity. Lentinan is a glucans as an immunostimulant [160]. These changes may indicate the
type of β-glucans with safe and pharmacological properties as well as way to a mortality rate reduction of fish species against various patho­
immunomodulatory, anti-oxidant, anti-inflammatory, anti-microbial, gens. Innovative β–glucan receptors namely Dectin-1 and Toll-like

300
S. Vijayaram et al. Fish and Shellfish Immunology 130 (2022) 294–308

receptors on leukocyte surfaces of vertebrates have provided various results through consumption; previous studies demonstrated that alginic
insights into the functions of immune responses induced by β-glucans acid could be used to enhance the nonspecific immune response in fish
[161]. Oral dietary administration of yeast-derived beta-glucans was species [125]. Dietary supplements of alginic acid (1 g/kg) improved
showed that numerous positive affects such as enhance phagocytosis, immune response, growth performance, and infection resistance against
lysozyme levels, respiratory burst activity, and resistance against bac­ WSSV disease in P. monodon, suitable dietary level of alginic acid is
terial pathogens in several cultured fish species. The function of glucans protecting immune system response against WSSV infection in juvenile
have been extensively studied in aquatic animals and the findings P. monodon [169]. Similarly, ergosan is an algal derivative with alginic
indicate that β -glucans improved growth and significantly reduced acid (2 g/kg), which could be used to improve innate immune response
mortality rate in certain species of aquatic animals [162,163]. β-glucans and infection resistance in different types of fish species [170]. Addi­
have been mentioned for their valuable role in several fish species such tionally, ergosan containing (0.5% and 1%) alginic acid derived from
as red sea bream (Pagrus major) [164], Atlantic Salmon (Salmo salar L.) is Laminaria digitata, could enhance the immune response, phagocytic ac­
utilized to improve growth, survival, immune system, disease resistance tivity, respiratory activity, and expression of interleukins and less
against aquatic pathogens and reduced mortality rate in aquatic animals response in a humoral immune parameter in rainbow trout (Onco­
[165]. rhynchus mykiss) [171]. The dietary additive of ergosan (5 g/kg)
The function of microbial-based bioactive compounds as immunos­ enhanced growth performance, protease, esterase activities, alkaline
timulants in aquatic animals has been summarised in Table 3. phosphatase, lysozyme actions, and disease resistance against Yersinia
ruckeri in rainbow trout (Oncorhynchus mykiss) (110 g) [172].
2.25. Algal bioactive immunostimulants
2.27. Fucoidan
2.25.1. Alginate
Alginate was derived from brown seaweed, and it is a polysaccharide Fucoidan is a complex long-chain sulfated polysaccharide, present in
component controlled by β-1, 4-D-mannuronic acid (M), and C5-epimer different species of brown algae. Commercially available fucoidan is
a-L-glucuronic acid (G). A dietary additive of sodium alginate was used usually derived from the different seaweed species namely Fucus ves­
to increase the non-specific immune system and infection resistance in iculosus, Cladosiphon okamuranus, Laminaria japonica, and Undaria pin­
Tilapia (Oreochromis niloticus) [125,166]. In addition, a nutritional ad­ natifida. Fucoidan (3% and 6%) promotes several biological effects like
ditive of sodium alginate (2 g/kg) enhanced the survival of grouper immunostimulation, disease resistance improvement, growth, and sur­
E. coioides by increasing its phagocytic activity, respiratory bursts, vival, whereas serum Albumin/Globulin (A/G) ratio and blood glucose
lysozyme activity, alternative complement activity, and SOD activity, level exhibited decreasing trend in diverse fishes and shrimps [173].
disease resistance against Streptococcus sp. and iridovirus infection in Saeed et al. (2021) reported that dietary fucoidan (200 mg/kg of body
grouper [167]. It has been reported that the nutritional additive of so­ weight, group 12–15 g) boosted phagocytic activity and disease resis­
dium alginate (2 g/kg) could improve phagocytic activity, respiratory tance of shrimps as well as overdoses of several immunostimulants to
bursts, lysozyme activity, alternative complement activity, SOD activity, induce immunosuppression in fish [174]. The side effects of immunos­
and disease resistance against V. alginolyticus but decreased GPX activity timulants have not been well-studied. Dietary supplements of fucoidan
in Pacific white shrimp (L. vannamei) [168]. (1 g/kg) increased the immune system (circulating hemocytes, PO ac­
tivity, RBs, phagocytic activity) and disease resistance against
V. alginolyticus in shrimps. The study reports concluded that dietary
2.26. Alginic acid fucoidan’s most important role is to enhance the immune system of
shrimp species [175]. Finally, oral administration of fucoidan (15 g/kg)
Alginic acid (or algin) is a naturally occurring, edible polysaccharide, derived from Sargassum polycystum could reduce the infection of White
present in brown algae. Such compound is hydrophilic and forms a Spot Syndrome Virus (WSSV) and reduce mortality rate, increase survival
viscous gum when hydrated. Accompanied by different metals such as level in black tiger shrimp P. monodon [176].
calcium and sodium, forms salts, which are recognized as alginates. Its
color ranges from white to yellowish-brown. Concerning its beneficial

Table 3
Microbial bioactive compoundsas immunostimulants in aquaculture.
Origin Bioactive Aquatic organisms Immune response References
immunostimulants

Microbes (Bacteria and Peptidoglycan kuruma shrimps ↑immune system response, RPA, antibacterial and antiviral effects [122,124,
fungi) Japanese flounder 125]
Penaeus monodon
sea cucumber Potential immunomodulatory factor [202]
Regulation of innate immune response
spotted sea bass [203]
Lipopolysaccharides Red sea bream ↑phagocytic activity and disease resistance against antibacterial activity [127,131]
trout
Channa argus Induced antioxidant and immune response [204]
Exopolysaccharides Zebrafish ↑immune system response, antibacterial effects, higher expression of TLR1 [189]
and TLR 2
Pacific white Good immune modulator and preventive factor against vibriosis [205]
shrimps
Zebrafish Modification changes of gut microbiota but not affected SCFAs [206]
Lipoteichoic Acid Silver pomfret ↑TLR, RPA [137]
Levan Labeo rohita ↑Lys, TP, A: G, RBA, RPA [140]
common carp Potent nutraceutical and immuno additive agents [207]
Cyprinus carpio Growth promoter and antibacterial agents [208]
Lentinan juvenile taimen ↑TGF-β, TNF-α, IL-1β, IL-6 and IL-8, ↑SOD, CAT, and GSH-Px, RPA [148,149]
Senegalese sole Modulate immune system and regulate gut microbiota [209]

↑- Enhance or Increase or Improve.

301
S. Vijayaram et al. Fish and Shellfish Immunology 130 (2022) 294–308

2.28. Laminarin expression and regulate immune pathways in flatfish (Senegalese sole)
juveniles. Dietary supplements of ulvans as sulfated polysaccharide el­
Laminarin is the most significant component derived from plants and ements contain numerous health-beneficial effects such as anticoagu­
seaweeds; Laminarin has a lot of bio-functional performances such as lant, antiviral, antioxidative, anticancer, and immunomodulating
antitumor, anti-apoptotic, anti-inflammatory, anticoagulant, and anti­ activities. In addition, dietary additives of ulvans help improve aquatic
oxidant activity. The biological activities of laminarin can be improved animal health and there is no negative impact on aquatic animals [186].
after appropriate chemical modifications, sulphation, and novel pro­ Dietary supplementation range of ulvans (0.05%) improved immuno­
cessing techniques. Studies on feeding laminarin-rich extracts to animals logical parameters (respiratory burst, total hemocyte count, and phe­
point out its suitability as an efficient component for food applications noloxidase activities) of shrimps and also survival and challenging tests
[177,178]. Laminarin is generally low molecular weight (e.g., 5 kDa) in giant tiger prawn (P. monodon) and Pacific white shrimp
and their inter-chain linkages with glucans residues are of great (L. vannamei), The dietary administration of ulvan was an effective
importance [179]. [180] Cui et al. (2021) demonstrated that laminarin immunostimulant for L. vannamei and P. monodon [187]. The function of
injection can change the expression of Arginine kinase (AK), and anti­ other original bioactive compounds as immunostimulants in aquatic
oxidant activity, and reduced the activity of Staphylococcus aureus, Lis­ animals has been summarised in Table 4.
teria monocytogenes, Escherichia coli, and Salmonella typhimurium.
Similarly [181], Shanmugavel and Krishnamoorthy (2021) reported that 3. Conclusion
laminarin may act as an immunostimulant and the energy source of
crustaceans [179]. Meng et al. (2020) demonstrated that dietary lami­ This review is based on the last few years’ literature data analysis and
narin of 1 g/kg enhanced growth activity, feed efficiency, and the provides an enlightening direction for bioactive immunostimulant
nonspecific immune reaction of E. coioides and also The levels of improvement in the aquaculture industry. As a comprehensive study
Creatinine (CREA) and UREA, as well as the action of alkaline phos­ was needed to improve bioactive immunostimulants, this review
phatase (ALP), were lower than of the control. There was no major concluded that bioactive immunostimulants sources, types, functions,
difference in the levels of alanine aminotransferase (ALT) and, aspartate and administration methods are beneficial for the improvement of
aminotransferase (AST) between control groups and treated groups. This healthy aquatic production and also for the increase of feed quality
study recommended that laminarin regulates the immune response and levels. Due to this review report, information was presented about
induces growth of the fish. bioactive immunostimulants, which are beneficial with a decreased
negative impact on aquatic animal health. Thus, such a report mainly
focused on positive responses analysis. The utilization of plant, animal,
2.29. Ulvans
bacterial, fungal, and algal-originated bioactive immunostimulants is
strongly recommended for improved aquatic animal health and aquatic
Ulvans are derived from the cell wall of green seaweeds; they contain
environment enhancement. Biomolecules’ functional feed additives
sulfated polysaccharides, Ulva was elevated in dietary fiber which
improve healthy aquatic productions and enhanced aquatic feed quality
stimulates gastrointestinal health and is linked to a reduction in the
levels and are also less toxic, cost-effective, and environmentally
incidence of chronic diseases as well as biological activities of dietary
friendly. The next steps of our research group will focus on the extrac­
ulvans contains several positive responses such as immunomodulating,
tion, characterization, and identification of biomolecules from different
antiviral, antioxidant, antihyperlipidemic and anticancer activity in
natural sources, biomolecules interaction analysis from the body, iden­
human and animals (including fish) [181,183]. In literature, it was re­
tification of novel bioactive immunostimulants, able to improve func­
ported for the first time that dietary supplementation (1 g/kg) of ulvans
tional nutritional supplements in the modern aquaculture industry. This
component could enhance immune system response and disease resis­
study information is enlightening to the future research direction of
tance against bacterial and parasitic infection and there was no signifi­
novel bioactive immunostimulants in the aquaculture industry.
cant difference in growth performance among fish for any of the diets
tested of Nile tilapia (Oreochromis niloticus) [184]. [185] Ponce et al.
(2020) suggested that U.ohnoi containing ulvans element was nontoxic Declaration of competing interest
and potential application as a vaccine adjuvant and/or nutraceuticals in
aquaculture. Ulvans (0.5 mg/fish) was used to enhance immune gene Authors have no conflict of interest to declare for the publication of

Table 4
Various bioactive compounds as immunostimulants in aquaculture.
Origin Bioactive immunostimulants Aquatic organisms Immune response References

Algae Alginic acid Penaeus monodonRainbow trout ↑ immune system, growth and antiviral activity [169,172]
Yeast β-Glucans Litopenaeusvannamei ↑phagocytic activity, RPA, resistance against bacterial pathogens [164,165]
Red sea bream
Atlantic Salmon
Mannanoligosaccharides Rainbow trout ↑TLC [154,156]
Nile tilapia ↑ blood protein, RPA
Seaweed Alginate Tilapia PA, PoA, RBA [166,167]
GrouperE. Coioides
Fucoidan P. monodon ↑ immune system, RPA [176]
Laminarin E. coioides ↑ non specific immunity, RPA [179]
Ulvans P. monodon THC, RBA, PoA [184,187]
L. vannamei ↑immune response, RPA
Nile tilapia

↑- Enhance or Increase or Improve.


Abbreviations of immunological parameters: ACP- Acid phosphatase; RPA-reduce pathogenic activity; IgM-ImmunoglobulinM; TLC-Total leukocyte count, THC-
Total haemocyte count; SOD-Superoxide dismutase; GSH-Px, Glutathione-peroxidase; CAT-Catalase; TAC - Total antioxidant capacity; Lys- Lysozyme activity; PA-
Phagocytic activity; PI- Phagocytic activity PoA-phenoloxidase activity; TP-Total protein; A:G, Albumin/Globulin; RBA- Respiratory burst activity; SOD- Super
oxide dismutase; RBC- Red blood cell; WBC - White blood cell; IL-1a; Interleukin 1a; TNF-a, Tumor Necrosis Factor a; TAS-Total antioxidant status; ACP- the alternative
complement pathway; PO activities-total peroxidase enzyme concentration; ACPA-the alternative complement pathway activity.

302
S. Vijayaram et al. Fish and Shellfish Immunology 130 (2022) 294–308

the present work. [18] M. Khodadadi, N. Abbasi, T.J. Adorian, H.G. Farsani, A. Hedayati, S.M. Hoseini,
Growth performance, survival, body composition, hematological parameters,
intestinal histomorphology, and digestive enzymes’ activity in juvenile rainbow
Data availability trout (Oncorhynchus mykiss) fed dietary Immunogen, J. Appl. Aquacult. 30 (2)
(2018) 174–186, https://doi.org/10.1080/10454438.2017.1420515.
Data will be made available on request. [19] R. Zhang, X.W. Wang, L.L. Liu, Y.C. Cao, H. Zhu, Dietary oregano essential oil
improved the immune response, activity of digestive enzymes, and intestinal
microbiota of the koi carp, Cyprinus carpio, Aquaculture 518 (2020), 734781,
Acknowledgments https://doi.org/10.1016/j.Aquaculture..2019.734781.
[20] M.E. Büyükdeveci, J.L. Balcázar, İ. Demirkale, S. Dikel, Effects of garlic-
supplemented diet on growth performance and intestinal microbiota of rainbow
We wish to acknowledge the national natural science foundation of trout (Oncorhynchus mykiss), Aquaculture 486 (2018) 170–174, https://doi.org/
China (Grant no. 32072990), Xiamen Marine and Fisheries Development 10.1016/j.Aquaculture..2017.12.022.
Fund (Grant No. 19CZP018HJ04), Industry-University Cooperation [21] F.J. Sutili, D.M. Gatlin III, B.M. Heinzmann, B. Baldisserotto, Plant essential oils
as fish diet additives: benefits on fish health and stability in feed, Rev. Aquacult.
Project of Fujian Province (Grant No. 2018N5011) for supporting this 10 (3) (2018) 716–726, https://doi.org/10.1111/raq.12197.
research work. This research work was partially supported by Chiang [22] E. Ringø, Z. Zhou, J.G. Vecino, S. Wadsworth, J. Romero, Å. Krogdahl, D.
Mai University, Chiang Mai, Thailand. L. Merrifield, Effect of dietary components on the gut microbiota of aquatic
animals. A never-ending story? Aquacult. Nutr. 22 (2) (2016) 219–282, https://
doi.org/10.1111/anu.12346.
References [23] X. Wu, T. Teame, Q. Hao, Q. Ding, H. Liu, C. Ran, Z. Zhang, Use of a paraprobiotic
and postbiotic feed supplement (HWF™) improves the growth performance,
[1] C.A. Wang, J. Li, L. Wang, Z. Zhao, L. Luo, X. Du, Q. Xu, Effects of dietary composition and function of gut microbiota in hybrid sturgeon (Acipenser baerii x
phosphorus on growth, body composition and immunity of young taimen Hucho Acipenser schrenckii), Fish Shellfish Immunol. 104 (2020) 36–45, https://doi.org/
taimen (Pallas, 1773), Aquacult. Res. 48 (6) (2017) 3066–3079, https://doi.org/ 10.1016/j.fsi.2020.05.054.
10.1111/are.13138. [24] S.H. Hoseinifar, A. Ahmadi, M. Raeisi, S.M. Hoseini, M. Khalili, N. Behnampour,
[2] C. Ragunath, V. Ramasubramanian, Dietary effect of padina boergesenii on Comparative study on immunomodulatory and growth enhancing effects of three
growth, immune response, and disease resistance against Pseudomonas aeruginosa prebiotics (galactooligosaccharide, fructooligosaccharide and inulin) in common
in Cirrhinus mrigala, Appl. Biochem. Biotechnol. 194 (5) (2022) 1881–1897. carp (Cyprinus carpio), Aquacult. Res. 48 (7) (2017) 3298–3307, https://doi.org/
https://link.springer.com/article/10.1007/s12010-021-03770-y. 10.1111/are.13156.
[3] J.P. Firmino, J. Galindo-Villegas, F.E. Reyes-López, E. Gisbert, Phytogenic [25] S.H. Hoseinifar, M. Dadar, E. Ringø, Modulation of nutrient digestibility and
bioactive compounds shape fish mucosal immunity, Front. Immunol. 12 (2021), digestive enzyme activities in aquatic animals: the functional feed additives
695973, https://doi.org/10.3389/fimmu.2021.695973. scenario, Aquacult. Res. 48 (8) (2017) 3987–4000, https://doi.org/10.1111/
[4] K. Yang, X. Qi, M. He, K. Song, F. Luo, X. Qu, F. Ling, Dietary supplementation of are.13368.
salidroside increases immune response and disease resistance of crucian carp [26] S.H. Hoseinifar, A. Mirvaghefi, M.A. Amoozegar, D.L. Merrifield, E. Ringø, In
(Carassius auratus) against Aeromonas hydrophila, Fish Shellfish Immunol. 106 vitro selection of a synbiotic and in vivo evaluation on intestinal microbiota,
(2020) 1–7, https://doi.org/10.1016/j.fsi.2020.07.054. performance and physiological response of rainbow trout (Oncorhynchus mykiss)
[5] T. Magrone, S. Fontana, F. Laforgia, T. Dragone, E. Jirillo, L. Passantino, fingerlings, Aquacult. Nutr. 23 (1) (2017) 111–118, https://doi.org/10.1111/
Administration of a polyphenol-enriched feed to farmed sea bass (Dicentrarchus anu.12373.
labrax L.) modulates intestinal and spleen immune responses, Oxid. Med. Cell. [27] S.H. Hoseinifar, R. Safari, M. Dadar, Dietary sodium propionate affects mucosal
Longev. (2016), https://doi.org/10.1155/2016/2827567, 2016. immune parameters, growth and appetite related genes expression: insights from
[6] T. Magrone, M.A. Russo, E. Jirillo, Dietary approaches to attain fish health with zebrafish model, Gen. Comp. Endocrinol. 243 (2017) 78–83, https://doi.org/
special reference to their immune system, Curr. Pharmaceut. Des. 24 (41) (2018) 10.1016/j.ygcen.2016.11.008.
4921–4931, https://doi.org/10.2174/1381612825666190104121544. [28] A. Rodríguez, J. Velázquez, L. González, T. Rodríguez-Ramos, B. Dixon, F.
[7] C. Zhou, H. Lin, X. Ge, J. Niu, J. Wang, Y. Wang, X. Tan, The effects of dietary H. Miyares, Y. Carpio, PACAP modulates the transcription of TLR-1/TLR-5/
soybean isoflavones on growth, innate immune responses, hepatic antioxidant MyD88 pathway genes and boosts antimicrobial defenses in Clarias gariepinus,
abilities and disease resistance of juvenile golden pompano Trachinotus ovatus, Fish Shellfish Immunol. 115 (2021) 150–159, https://doi.org/10.1016/j.
Fish Shellfish Immunol. 43 (1) (2015) 158–166, https://doi.org/10.1016/j. fsi.2021.06.009.
fsi.2014.12.014. [29] S.H. Hoseinifar, G. Rashidian, H. Ghafarifarsani, M.A. Jahazi, M. Soltani, H.
[8] G. Ren, K. Li, Y. Hu, M. Yu, J. Qu, X. Xu, Optimization of selenizing conditions for V. Doan, M. Paolucci, Effects of apple (Malus pomila) pomace-derived pectin on
Seleno-Lentinan and its characteristics, Int. J. Biol. Macromol. 81 (2015) the innate immune responses, expressions of key immune-related genes, growth
249–258, https://doi.org/10.1016/j.ijbiomac.2015.08.012. performance, and digestive enzyme activity of rainbow trout (Oncorhynchus
[9] N. Sadat Hoseini Madani, T.J. Adorian, H. Ghafari Farsani, S.H. Hoseinifar, The mykiss), Animals 11 (7) (2021) 2117. https://www.mdpi.com/2076-2615/11/7/
effects of dietary probiotic Bacilli (Bacillus subtilis and Bacillus licheniformis) on 2117.
growth performance, feed efficiency, body composition and immune parameters [30] U.S. Sagaram, M.S. Gaikwad, R. Nandru, S. Dasgupta, Microalgae as feed
of whiteleg shrimp (Litopenaeus vannamei) postlarvae, Aquacult. Res. 49 (5) ingredients: recent developments on their role in immunomodulation and gut
(2018) 1926–1933, https://doi.org/10.1111/are.13648. microbiota of aquaculture species, FEMS (Fed. Eur. Microbiol. Soc.) Microbiol.
[10] S.H. Hoseinifar, Y.Z. Sun, Z. Zhou, H. Van Doan, S.J. Davies, R. Harikrishnan, Lett. 368 (11) (2021), https://doi.org/10.1093/femsle/fnab071 fnab071.
Boosting immune function and disease bio-control through environment-friendly [31] M.F.Z. Ali, S. Nakahara, Y. Otsu, A. Ido, C. Miura, T. Miura, Effects of functional
and sustainable approaches in finfish aquaculture: herbal therapy scenarios, Rev. polysaccharide from silkworm as an immunostimulant on transcriptional
Fish. Sci. Aquacul. 28 (3) (2020) 303–321, https://doi.org/10.1080/ profiling and disease resistance in fish, J. Insects as Food and Feed (2021) 1–14,
23308249.2020.1731420. https://doi.org/10.3920/JIFF2021.0108.
[11] O. Alfred, A. Shaahu, D.A. Orban, M. Egwenomhe, An overview on understanding [32] V.K. Gurjar, D. Pal, Natural compounds extracted from medicinal plants and their
the basic concept of fish diseases in aquaculture, IRE J. 4 (6) (2020). http immunomodulatory activities, in: Bioactive Natural Products for Pharmaceutical
s://www.irejournals.com/formatedpaper/1702541.pdf. Applications, Springer, Cham, 2021, pp. 197–261, https://doi.org/10.1007/978-
[12] J.A. Gephart, C.D. Golden, F. Asche, B. Belton, C. Brugere, H.E. Froehlich, E. 3-030-54027-2_6.
H. Allison, Scenarios for global aquaculture and its role in human nutrition, Rev. [33] M.G. Mendoza Rodriguez, C. Pohlenz, D.M. Gatlin III, Supplementation of organic
Fish. Sci. Aquacul. 29 (1) (2020) 122–138, https://doi.org/10.1080/ acids and algae extracts in the diet of red drum Sciaenops ocellatus: immunological
23308249.2020.1782342. impacts, Aquacult. Res. 48 (4) (2017) 1778–1786, https://doi.org/10.1111/
[13] R.N. Roy, Bioactive natural derivatives of phthalate ester, Crit. Rev. Biotechnol. are.13015.
40 (7) (2020) 913–929, https://doi.org/10.1080/07388551.2020.1789838. [34] S. Luna-Castro, I. Ceballos-Olvera, F. Benavides-González, Z. Blanco-Martínez,
[14] G. Wu, Nutrition and metabolism: foundations for animal growth, development, G. Sánchez-Martínez, M.D.L.L. Vázquez-Sauceda, M. de la Garza, Bovine
reproduction, and health, in: Recent Advances in Animal Nutrition and lactoferrin in fish culture: current research and future directions, Aquacult. Res.
Metabolism, Springer, Cham, 2022, pp. 1–24, https://doi.org/10.1007/978-3- 53 (3) (2022) 735–745, https://doi.org/10.1111/are.15621.
030-85686-1_1. [35] H. Minářová, L. Bláhová, J. Kalina, I. Papežíková, J. Mareš, R. Kopp, M. Palíková,
[15] V. Šimat, N. Elabed, P. Kulawik, Z. Ceylan, E. Jamroz, H. Yazgan, F. Özogul, Plant-based and immunostimulant-enhanced diets modulate oxidative stress,
Recent advances in marine-based nutraceuticals and their health benefits, Mar. immune and haematological indices in rainbow trout (Oncorhynchus mykiss), Acta
Drugs 18 (12) (2020) 627, https://doi.org/10.3390/md18120627. Vet. 90 (2) (2021) 233–253. https://actavet.vfu.cz/90/2/233/.
[16] S.A. Ashraf, M. Adnan, M. Patel, A.J. Siddiqui, M. Sachidanandan, M. Snoussi, [36] C. Lumsangkul, W. Tapingkae, K. Sringarm, S. Jaturasitha, C. Le Xuan,
S. Hadi, Fish-based bioactives as potent nutraceuticals: exploring the therapeutic S. Wannavijit, H. Van Doan, Effect of dietary sugarcane bagasse supplementation
perspective of sustainable food from the sea, Mar. Drugs 18 (5) (2020) 265, on growth performance, immune response, and immune and antioxidant-related
https://doi.org/10.3390/md18050265. gene expressions of Nile tilapia (Oreochromis niloticus) cultured under biofloc
[17] P. Encarnação, Functional feed additives in aquaculture feeds, in: Aquafeed system, Animals 11 (7) (2021) 2035, https://doi.org/10.3390/ani11072035.
Formulation, Academic Press, 2016, pp. 217–237, https://doi.org/10.1016/ [37] P. Yarahmadi, H.G. Farsani, A. Khazaei, M. Khodadadi, G. Rashidiyan, M.
B978-0-12-800873-7.00005-1. A. Jalali, Protective effects of the prebiotic on the immunological indicators of
rainbow trout (Oncorhynchus mykiss) infected with Aeromonas hydrophila, Fish

303
S. Vijayaram et al. Fish and Shellfish Immunology 130 (2022) 294–308

Shellfish Immunol. 54 (2016) 589–597, https://doi.org/10.1016/j. [58] Q. Ye, Y. Feng, Z. Wang, A. Zhou, S. Xie, L. Fan, J. Zou, Effects of dietary
fsi.2016.05.010. Gelsemium elegans alkaloids on intestinal morphology, antioxidant status,
[38] R.B. Dixit, U.S. Sagaram, C. Gocher, G.R. Krishna Kumar, S. Dasgupta, immune responses and microbiota of Megalobrama amblycephala, Fish Shellfish
Biomolecular characterisation of marine microalga in comparison to fishmeal and Immunol. 94 (2019) 464–478, https://doi.org/10.1016/j.fsi.2019.09.048.
soymeal as an alternative feed ingredient, Phytochem. Anal. 33 (3) (2022) [59] X. Zhou, Y. Peng, L. Li, K. He, T. Huang, S. Mou, X. Li, Effects of dietary
365–372, https://doi.org/10.1002/pca.3094. supplementations with the fibrous root of Rhizoma Coptidis and its main
[39] M. Abha, S. Abhisweta, Effect of curcumin, ascorbic acid against chlorpyrifos alkaloids on non-specific immunity and disease resistance of common carp, Vet.
toxicity on histotexture and bimolecular content of fresh water catfish, Immunol. Immunopathol. 173 (2016) 34–38, https://doi.org/10.1016/j.
Heteropneustes fossilis, J. Exp. Zool. India 23 (2) (2020) 1043–1049. https:// vetimm.2016.03.014.
www.cabdirect.org/cabdirect/abstract/20203390895. [60] M. Kherade, S. Solanke, M. Tawar, S. Wankhede, Fructooligosaccharides: a
[40] F. Baleta, P. Candelaria, D.E. Baldo, L.J. Baleta, L. Plantado, M. Navarro, Dietary comprehensive review, J. Ayu. Herb. Med 7 (2021) 193–200, https://doi.org/
incorporation of Sweet Potato Ipomoea batatas shoots improved growth 10.31254/jahm.2021.7305.
performance and haematological profile of Tilapia Oreochromis niloticus in Hapa [61] A. Nawaz, S. Irshad, S.H. Hoseinifar, H. Xiong, The functionality of prebiotics as
Nets, Aquat. Res. 5 (1) (2022) 1–10, https://doi.org/10.3153/AR22001. immunostimulant: evidences from trials on terrestrial and aquatic animals, Fish
[41] M.T. El-Saadony, N.M. Zabermawi, N.M. Zabermawi, M.A. Burollus, M.E. Shafi, Shellfish Immunol. 76 (2018) 272–278, https://doi.org/10.1016/j.
M. Alagawany, M.E. Abd El-Hack, Nutritional aspects and health benefits of fsi.2018.03.004.
bioactive plant compounds against infectious diseases: a review, Food Rev. Int. [62] D.B. Bebo, J. Zhu, S.M. Sun, C.Y. Song, Effects of Antibiotics on the Growth,
1–23 (2021), https://doi.org/10.1080/87559129.2021.1944183. Antioxidant Capacity, Activity of Digestive Enzymes and Serum Biochemical
[42] M. Paolucci, M.G. Volpe, S. Costantini, Polyphenol enriched diets for aquaculture Indices, Liver and Intestine Morphology of Megalobrama amblycephala Fry, 2020.
applications, J. Int. Soc. Antioxid. Nutr. Health 3 (4) (2016). https://www.fisheriesjournal.com/archives/2020/vol8issue3/PartG/8-3-39-522.
[43] M.A. Dawood, S. Koshio, M.Á. Esteban, Beneficial roles of feed additives as pdf.
immunostimulants in aquaculture: a review, Rev. Aquacult. 10 (4) (2018) [63] H. Ghafarifarsani, S.H. Hoseinifar, M. Aftabgard, H. Van Doan, The improving
950–974, https://doi.org/10.1111/raq.12209. role of savory (Satureja hortensis) essential oil for Caspian roach (Rutilus caspicus)
[44] M. Arciuli, D. Fiocco, S. Fontana, M.P. Arena, M.A. Frassanito, A. Gallone, fry: growth, haematological, immunological, and antioxidant parameters and
Administration of a polyphenol-enriched feed to farmed sea bass (Dicentrarchus resistance to salinity stress, Aquaculture 548 (2022), 737653, https://doi.org/
labrax L.): kidney melanomacrophages response, Fish Shellfish Immunol. 68 10.1016/j.Aquaculture..2021.737653.
(2017) 404–410, https://doi.org/10.1016/j.fsi.2017.07.043. [64] M.T. Hasan, W. Je Jang, J.M. Lee, B.J. Lee, S.W. Hur, S. Gu Lim, I.S. Kong, Effects
[45] M. Long, W. Lin, J. Hou, H. Guo, L. Li, D. Li, F. Yang, Dietary supplementation of immunostimulants, prebiotics, probiotics, synbiotics, and potentially
with selenium yeast and tea polyphenols improve growth performance and nitrite immunoreactive feed additives on olive flounder (Paralichthys olivaceus): a
tolerance of Wuchang bream (Megalobrama amblycephala), Fish Shellfish review, Rev. Fish. Sci. Aquacul. 27 (4) (2019) 417–437, https://doi.org/10.1080/
Immunol. 68 (2017) 74–83, https://doi.org/10.1016/j.fsi.2017.07.017. 23308249.2019.1622510.
[46] M.G. Volpe, S. Costantini, E. Coccia, L. Parrillo, M. Paolucci, Evaluation of [65] L. Poolsawat, X. Li, H. Yang, P. Yang, M.A. Kabir Chowdhury, A. Yusuf, X. Leng,
metabolic changes induced by polyphenols in the crayfish Astacus leptodactylus The potentials of fructooligosaccharide on growth, feed utilization, immune and
by metabolomics using Fourier transformed infrared spectroscopy, J. Biosci. 43 antioxidant parameters, microbial community and disease resistance of tilapia
(4) (2018) 585–596, https://doi.org/10.1007/s12038-018-9774-1. (Oreochromis niloticus× O. aureus), Aquacult. Res. 51 (11) (2020) 4430–4442,
[47] H. Van Doan, S.H. Hoseinifar, K. Sringarm, S. Jaturasitha, B. Yuangsoi, M. https://doi.org/10.1111/are.14786.
A. Dawood, C. Faggio, Effects of Assam tea extract on growth, skin mucus, serum [66] A. Raza, S. Iqbal, Z. Ahmad, M.A. Rehman, M. Waseem, M. Usman, Conversion of
immunity and disease resistance of Nile tilapia (Oreochromis niloticus) against milk lactose to galacto-oligosaccharides by enzymes to produce prebiotic
Streptococcus agalactiae, Fish Shellfish Immunol. 93 (2019) 428–435, https:// enriched cheese, Future Foods 4 (2021), 100097, https://doi.org/10.1016/j.
doi.org/10.1016/j.fsi.2019.07.077. fufo.2021.100097.
[48] T. Magrone, A. Spagnoletta, M. Magrone, M.A. Russo, A. Corriero, E. Jirillo, [67] A. Rashidinejad, A. Bahrami, A. Rehman, A. Rezaei, A. Babazadeh, H. Singh, S.
L. Passantino, Effects of polyphenol administration to European farmed sea bass M. Jafari, Co-encapsulation of probiotics with prebiotics and their application in
(Dicentrharcus labrax L.): special focus on hepatopancreas morphology, Endocr. functional/synbiotic dairy products, Crit. Rev. Food Sci. Nutr. 62 (9) (2022)
Metab. Immune Disord. - Drug Targets 19 (4) (2019) 526–533, https://doi.org/ 2470–2494. https://www.tandfonline.com/doi/abs/10.1080/10408398.2020
10.2174/1871530318666181009111214. .1854169.
[49] L. Zhong, Y. Hu, Y. Hu, J. Li, Y. Tian, J. Chen, T. Xiao, Effects of dietary tea [68] S.H. Hoseinifar, F. Zoheiri, M. Dadar, R. Rufchaei, E. Ringø, Dietary
polyphenols on growth, immunity and lipid metabolism of juvenile black carp galactooligosaccharide elicits positive effects on non-specific immune parameters
Mylopharyngodon piceus, Aquacult. Res. 51 (2) (2020) 569–576, https://doi.org/ and growth performance in Caspian white fish (Rutilus frisii kutum) fry, Fish
10.1111/are.14403. Shellfish Immunol. 56 (2016) 467–472, https://doi.org/10.1016/j.
[50] R. Safari, S.H. Hoseinifar, M.R. Imanpour, M. Mazandarani, H. Sanchouli, fsi.2016.08.001.
M. Paolucci, Effects of dietary polyphenols on mucosal and humoral immune [69] M.T. Hasan, W.J. Jang, H. Kim, B.J. Lee, K.W. Kim, S.W. Hur, I.S. Kong,
responses, antioxidant defense and growth gene expression in beluga sturgeon Synergistic effects of dietary Bacillus sp. SJ-10 plus β-glucooligosaccharides as a
(Huso huso), Aquaculture 528 (2020), 735494, https://doi.org/10.1016/j. synbiotic on growth performance, innate immunity and streptococcosis resistance
Aquaculture..2020.735494. in olive flounder (Paralichthys olivaceus), Fish Shellfish Immunol. 82 (2018)
[51] M.A. Jahazi, S.H. Hoseinifar, V. Jafari, A. Hajimoradloo, H. Van Doan, 544–553, https://doi.org/10.1016/j.Aquaculture..2017.09.020.
M. Paolucci, Dietary supplementation of polyphenols positively affects the innate [70] K. Yukgehnaish, P. Kumar, P. Sivachandran, K. Marimuthu, A. Arshad, B.A. Paray,
immune response, oxidative status, and growth performance of common carp, J. Arockiaraj, Gut microbiota metagenomics in aquaculture: factors influencing
Cyprinus carpio L, Aquaculture 517 (2020), 734709, https://doi.org/10.1016/j. gut microbiome and its physiological role in fish, Rev. Aquacult. 12 (3) (2020)
Aquaculture..2019.734709. 1903–1927, https://doi.org/10.1111/raq.12416.
[52] S.M. Hussain, H. Gohar, M. Asrar, M.M. Shahzad, A. Rasul, M. Hussain, A. Sharif, [71] R. Tarkhani, A. Imani, S.H. Hoseinifar, O. Ashayerizadeh, K.S. Moghanlou,
Effects of Polyphenols Supplemented Canola Meal Based Diet on Proximate R. Manaffar, M. Reverter, Comparative study of host-associated and commercial
Composition, Minerals Absorption and Hematology of Cyprinus carpio probiotic effects on serum and mucosal immune parameters, intestinal
Fingerlings, 2021. https://researcherslinks.com/uploads/articles/1620236631 microbiota, digestive enzymes activity and growth performance of roach (Rutilus
PJZ_MH20190214170258-R3_Hussain%20et%20al.pdf. rutilus caspicus) fingerlings, Fish Shellfish Immunol. 98 (2020) 661–669, https://
[53] U. Sarker, S. Oba, Polyphenol and flavonoid profiles and radical scavenging doi.org/10.1016/j.fsi.2019.10.063.
activity in leafy vegetable Amaranthus gangeticus, BMC Plant Biol. 20 (1) (2020) [72] S.H. Hoseinifar, A. Mirvaghefi, M.A. Amoozegar, M. Sharifian, M.Á. Esteban,
1–12, https://doi.org/10.1186/s12870-020-02700-0. Modulation of innate immune response, mucosal parameters and disease
[54] M. Li, X. Zhu, J. Tian, M. Liu, G. Wang, Dietary flavonoids from Allium resistance in rainbow trout (Oncorhynchus mykiss) upon synbiotic feeding, Fish
mongolicum Regel promotes growth, improves immune, antioxidant status, Shellfish Immunol. 45 (1) (2015) 27–32, https://doi.org/10.1016/j.
immune-related signaling molecules and disease resistance in juvenile northern fsi.2015.03.029.
snakehead fish (Channa argus), Aquaculture 501 (2019) 473–481, https://doi. [73] M. Modanloo, S. Soltanian, M. Akhlaghi, S.H. Hoseinifar, The effects of single or
org/10.1016/j.Aquaculture..2018.12.011. combined administration of galactooligosaccharide and Pediococcus acidilactici on
[55] Z.G. Sun, Z.N. Li, X.W. Miao, S. Li, H.L. Zhu, Recent advances in natural products cutaneous mucus immune parameters, humoral immune responses and immune
with antiviral activities, Mini Rev. Med. Chem. 21 (14) (2021) 1888–1908, related genes expression in common carp (Cyprinus carpio) fingerlings, Fish
https://doi.org/10.2174/1389557521666210304110824. Shellfish Immunol. 70 (2017) 391–397, https://doi.org/10.1016/j.
[56] C.G. Awuchi, The biochemistry, toxicology, and uses of the pharmacologically fsi.2017.09.032.
active phytochemicals: alkaloids, terpenes, polyphenols, and glycosides, J. Food [74] S.H. Hoseinifar, F. Zoheiri, C.M. Caipang, Dietary sodium propionate improved
Pharm. Sci. (2019) 2. -2, file:///C:/Users/Hii/AppData/Local/Temp/666-Article performance, mucosal and humoral immune responses in Caspian white fish
%20Text-2613-4-10-20210723.pdf. (Rutilus frisii kutum) fry, Fish Shellfish Immunol. 55 (2016) 523–528, https://doi.
[57] S. Andayani, M. Fadjar, A.P. Rahayu, M.F. Rahman, E. Kurniati, Potency of org/10.1016/j.fsi.2016.06.027.
jellyfish alkaloid (Aeginura sp) on kidney histopathology and relative percent [75] O. Safari, M. Paolucci, Modulation of growth performance, immunity, and disease
survival (RPS) of tiger grouper (Epinephelus fuscoguttatus) against Vibrio resistance in narrow-clawed crayfish, Astacus leptodactylus leptodactylus
harveyi, No. 1, in: IOP Conference Series: Earth and Environmental Science, vol. (Eschscholtz, 1823) upon synbiotic feeding, Aquaculture 479 (2017) 333–341,
584, 2020, October, 12065. IOP Publishing, https://iopscience.iop.org/article/1 https://doi.org/10.1016/j.Aquaculture..2017.05.049.
0.1088/1755-1315/584/1/012065/meta. [76] H. Ghafarifarsani, G. Rashidian, T. Bagheri, S.H. Hoseinifar, H. Van Doan, Study
on growth enhancement and the protective effects of dietary prebiotic inulin on

304
S. Vijayaram et al. Fish and Shellfish Immunology 130 (2022) 294–308

immunity responses of rainbow trout fry infected with, Ann. Anim. Sci. 21 (2) [98] R. Sowmya, N.M. Sachindra, Enhancement of non-specific immune responses in
(2021) 543–559, https://doi.org/10.2478/aoas-2020-0074. common carp, Cyprinus carpio, by dietary carotenoids obtained from shrimp
[77] S.K. Gupta, B. Sarkar, M. Priyam, N. Kumar, S. Naskar, M.J. Foysal, T.R. Sharma, exoskeleton, Aquacult. Res. 46 (7) (2015) 1562–1572, https://doi.org/10.1111/
Inflammatory and stress biomarker response of Aeromonas hydrophila infected are.12310.
rohu, Labeo rohita fingerlings to dietary microbial levan, Aquaculture 521 (2020), [99] T. Nakano, G. Wiegertjes, Properties of carotenoids in fish fitness: a review, Mar.
735020, https://doi.org/10.1016/j.Aquaculture..2020.735020. Drugs 18 (11) (2020) 568, https://doi.org/10.3390/md18110568.
[78] W.Y. Mo, Z. Cheng, W.M. Choi, C.H. Lun, Y.B. Man, J.T. Wong, M.H. Wong, Use of [100] S.K. Singh, M.P. Bhandari, P. Timalsina, Effect of dietary supplementation of
food waste as fish feeds: effects of prebiotic fibers (inulin and carotenoids on growth performance of rainbow trout (Oncorhynchus mykiss,
mannanoligosaccharide) on growth and non-specific immunity of grass carp Walbaum) in Rasuwa, Nepal. Nepalese J. Agric. Sci, 75, https://hicast.edu.
(Ctenopharyngodon idella), Environ. Sci. Pollut. Control Ser. 22 (22) (2015) np/wp-ntent/uploads/2021/08/NJAS_2021_V21_July.pdf#page=76.
17663–17671. https://link.springer.com/article/10.1007/s11356-015-4971-z. [101] J.M. Joy, A. Joseph, R. Anandan, The Role of Carotenoids in Enhancing the
[79] S. Syed Raffic Ali, K. Ambasankar, P.E. Praveena, S. Nandakumar, M. Saiyad Health of Aquatic Organisms, 2021. http://krishi.icar.gov.in/jspui/han
Musthafa, Effect of dietary prebiotic inulin on histology, immuno-haematological dle/123456789/60365.
and biochemical parameters of Asian seabass (Lates calcarifer), Aquacult. Res. 49 [102] M.S. Hassaan, E.Y. Mohammady, M.R. Soaudy, S.A. Sabae, A.M. Mahmoud, E.
(8) (2018) 2732–2740, https://doi.org/10.1111/are.13734. R. El-Haroun, Comparative study on the effect of dietary β-carotene and
[80] X. Zhang, L. Xie, J. Long, Q. Xie, Y. Zheng, K. Liu, X. Li, Salidroside: a review of its phycocyanin extracted from Spirulina platensis on immune-oxidative stress
recent advances in synthetic pathways and pharmacological properties, Chem. biomarkers, genes expression and intestinal enzymes, serum biochemical in Nile
Biol. Interact. 339 (2021), 109268, https://doi.org/10.1016/j.cbi.2020.109268. tilapia, Oreochromis niloticus, Fish Shellfish Immunol. 108 (2021) 63–72,
[81] M. Zare, M.K. Mirzakhani, V. Stejskal, Growth, body proximate composition and https://doi.org/10.1016/j.fsi.2020.11.012.
selected blood parameters of rainbow trout Onchorhynchus mykiss fingerlings fed [103] A. Babin, C. Saciat, M. Teixeira, J.P. Troussard, S. Motreuil, J. Moreau, Y. Moret,
on a diet supplemented with nettle Urtica dioica and tarragon Artemisia Limiting immunopathology: interaction between carotenoids and enzymatic
dracunculus, Aquacult. Res. 52 (11) (2021) 5691–5702, https://doi.org/ antioxidant defences, Dev. Comp. Immunol. 49 (2) (2015) 278–281, https://doi.
10.1111/are.15444. org/10.1016/j.dci.2014.12.007.
[82] J. Xie, H. Fang, X. He, S. Liao, Y. Liu, L. Tian, J. Niu, Study on mechanism of [104] X. Yuan, J. Zheng, S. Jiao, G. Cheng, C. Feng, Y. Du, H. Liu, A review on the
synthetic astaxanthin and Haematococcus pluvialis improving the growth preparation of chitosan oligosaccharides and application to human health, animal
performance and antioxidant capacity under acute hypoxia stress of golden husbandry and agricultural production, Carbohydr. Polym. 220 (2019) 60–70,
pompano (Trachinotus ovatus) and enhancing anti-inflammatory by activating https://doi.org/10.1016/j.carbpol.2019.05.050.
Nrf2-ARE pathway to antagonize the NF-κB pathway, Aquaculture 518 (2020), [105] D. Kamilya, M.I.R. Khan, Chitin and chitosan as promising immunostimulant for
734657, https://doi.org/10.1016/j.Aquaculture..2019.734657. aquaculture, in: Handbook of Chitin and Chitosan, Elsevier, 2020, pp. 761–771,
[83] L. Chen, M. Li, Z. Yang, W. Tao, P. Wang, X. Tian, W. Wang, Gardenia jasminoides https://doi.org/10.1016/B978-0-12-817966-6.00024-8.
Ellis: ethnopharmacology, phytochemistry, and pharmacological and industrial [106] I.V. Zadinelo, L.D. dos Santos, H.J. Alves, A. de Marco Viott, L. de Souza Neves
applications of an important traditional Chinese medicine, J. Ethnopharmacol. Ellendersen, G.I.B. de Muniz, R.A. Bombardelli, Chitosan foam–based filter:
257 (2020), 112829, https://doi.org/10.1016/j.jep.2020.112829. maintenance of water quality for nile Tilapia cultivation, Water, Air, Soil Pollut.
[84] W. Sun, X. Li, H. Xu, J. Chen, X. Xu, X. Leng, Effects of dietary geniposide on 231 (10) (2020) 1–20, https://doi.org/10.1007/s11270-020-04905-3.
growth, flesh quality, and lipid metabolism of grass carp, Ctenopharyngodon idella, [107] W. Wang, C. Xue, X. Mao, Chitosan: structural modification, biological activity
J. World Aquacult. Soc. 48 (6) (2017) 927–937, https://doi.org/10.1111/ and application, Int. J. Biol. Macromol. 164 (2020) 4532–4546, https://doi.org/
jwas.12412. 10.1016/j.ijbiomac.2020.09.042.
[85] A.G. Huang, X. Tu, X.Z. Qi, F. Ling, B. Zhu, G.X. Wang, Gardenia jasminoides Ellis [108] S. Saini, A. Dhiman, S. Nanda, Immunomodulatory properties of chitosan: impact
inhibit white spot syndrome virus replication in red swamp crayfish Procambarus on wound healing and tissue repair, Endocr. Metab. Immune Disord. - Drug
clarkii, Aquaculture 504 (2019) 239–247, https://doi.org/10.1016/j. Targets 20 (10) (2020) 1611–1623, https://doi.org/10.2174/
Aquaculture..2019.02.008. 1871530320666200503054605.
[86] E. Wang, X. Chen, K. Wang, J. Wang, D. Chen, Y. Geng, X. Wei, Plant [109] D.M. Gatlin III, F.Y. Yamamoto, Nutritional supplements and fish health, in: Fish
polysaccharides used as immunostimulants enhance innate immune response and Nutrition, Academic Press, 2022, pp. 745–773, https://doi.org/10.1016/B978-0-
disease resistance against Aeromonas hydrophila infection in fish, Fish Shellfish 12-819587-1.00004-5.
Immunol. 59 (2016) 196–202, https://doi.org/10.1016/j.fsi.2016.10.039. [110] H.M. Abdel-Ghany, M.E.S. Salem, Effects of dietary chitosan supplementation on
[87] M. He, G. Liu, Y. Liu, K. Yang, X. Qi, A. Huang, E. Wang, Effects of geniposide as farmed fish; a review, Rev. Aquacult. 12 (1) (2020) 438–452, https://doi.org/
immunostimulant on the innate immune response and disease resistance in 10.1111/raq.12326.
crucian carp, Aquaculture 529 (2020), 735713, https://doi.org/10.1016/j. [111] A. Gupta, S.K. Gupta, M. Priyam, M.A. Siddik, N. Kumar, P.K. Mishra,
Aquaculture..2020.735713. A. Pattanayak, Immunomodulation by dietary supplements: a preventive health
[88] M. Tsaneva, E.J. Van Damme, 130 years of plant lectin research, Glycoconj. J. 37 strategy for sustainable aquaculture of tropical freshwater fish, Labeo rohita
(5) (2020) 533–551, https://doi.org/10.1007/s10719-020-09942-y. (Hamilton, 1822), Rev. Aquacult. 13 (4) (2021) 2364–2394, https://doi.org/
[89] Y. Huang, F. Jiang, R. Wang, Y. Shi, S. Hu, T. Wu, Z. Zhao, In silico 10.1111/raq.12581.
characterization and expression analysis of eight C-type lectins in obscure puffer [112] A.K. Oushani, M. Soltani, N. Sheikhzadeh, M.S. Mehrgan, H.R. Islami, Effects of
Takifugu obscurus, Vet. Immunol. Immunopathol. 234 (2021), 110200, https:// dietary chitosan and nano-chitosan loaded clinoptilolite on growth and immune
doi.org/10.1016/j.vetimm.2021.110200. responses of rainbow trout (Oncorhynchus mykiss), Fish Shellfish Immunol. 98
[90] Y. Huang, Y. Shi, S. Hu, T. Wu, Z. Zhao, Characterization and functional analysis (2020) 210–217, https://doi.org/10.1016/j.fsi.2020.01.016.
of two transmembrane C-type lectins in obscure puffer (Takifugu obscurus), Front. [113] Y. Wu, A. Rashidpour, M.P. Almajano, I. Metón, Chitosan-based drug delivery
Immunol. 11 (2020) 436, https://doi.org/10.3389/fimmu.2020.00436. system: applications in fish biotechnology, Polymers 12 (5) (2020) 1177, https://
[91] D.L. Lindenwald, B. Lepenies, C-Type lectins in veterinary species: recent doi.org/10.3390/polym12051177.
advancements and applications, Int. J. Mol. Sci. 21 (14) (2020) 5122, https://doi. [114] H. Mondal, N. Chandrasekaran, A. Mukherjee, J. Thomas, Viral infections in
org/10.3390/ijms21145122. cultured fish and shrimps: current status and treatment methods, Aquacult. Int.
[92] S. Narla, M. Azzam, S. Townsend, G. Vellaichamy, A.V. Marzano, A. Alavi, I. (2021) 1–36. https://link.springer.com/article/10.1007/s10499-021-00795-2.
H. Hamzavi, Identifying key components and therapeutic targets of the immune [115] N.M. Abu-Elala, S.H. Mohamed, M.M. Zaki, A.E. Eissa, Assessment of the immune-
system in hidradenitis suppurativa with an emphasis on neutrophils, Br. J. modulatory and antimicrobial effects of dietary chitosan on Nile tilapia
Dermatol. 184 (6) (2021) 1004–1013, https://doi.org/10.1111/bjd.19538. (Oreochrmis niloticus) with special emphasis to its bio-remediating impacts, Fish
[93] V. Morales-Moya, F. Stambuk, R. Farlora, F. Guzmán, L. Mercado, K. Brokordt, Shellfish Immunol. 46 (2) (2015) 678–685, https://doi.org/10.1016/j.
P. Schmitt, Molecular characterization and cellular localization of a fsi.2015.08.004.
transmembrane C-type lectin receptor in hemocytes from the scallop Argopecten [116] B. Wang, Y.P. Timilsena, E. Blanch, B. Adhikari, Lactoferrin: structure, function,
purpuratus, Aquaculture 546 (2022), 737293, https://doi.org/10.1016/j. denaturation and digestion, Crit. Rev. Food Sci. Nutr. 59 (4) (2019) 580–596,
Aquaculture..2021.737293. https://doi.org/10.1080/10408398.2017.1381583.
[94] K. Zhang, X. Liu, X. Li, Y. Liu, H. Yu, J. Liu, Q. Zhang, Antibacterial functions of a [117] P. Elumalai, A. Kurian, S. Lakshmi, C. Faggio, M.A. Esteban, E. Ringø, Herbal
novel fish-egg lectin from spotted knifejaw (Oplegnathus punctatus) during host immunomodulators in aquaculture, Rev. Fish. Sci. Aquacul. 29 (1) (2020) 33–57,
defense immune responses, Dev. Comp. Immunol. 111 (2020), 103758, https:// https://doi.org/10.1080/23308249.2020.1779651.
doi.org/10.1016/j.dci.2020.103758. [118] V. Morshedi, N. Agh, F. Noori, F. Jafari, A. Ghasemi, M.T. Mozanzadeh, Effects of
[95] E. Catanzaro, C. Calcabrini, A. Bishayee, C. Fimognari, Antitumor potential of single and combined supplementation of dietary probiotic with bovine lactoferrin
marine and freshwater lectins, Mar. Drugs 18 (1) (2019) 11, https://doi.org/ and xylooligosaccharide on hemato-immunological and digestive enzymes of
10.3390/md18010011. silvery-black porgy fingerlings, Ann. Anim. Sci. 20 (1) (2020) 137–155. https://
[96] W. Liang, R. Wu, T. Yang, H. Shen, Z. Hu, Effect of pathogenic bacteria on a novel sciendo.com/downloadpdf/journals/aoas/20/1/article-p137.xml.
C-type lectin, hemocyte and superoxide dismutase/alkaline phosphatase activity [119] K.E. Fernandes, D.A. Carter, The antifungal activity of lactoferrin and its derived
in Onchidium reevesii, Fish Shellfish Immunol. 102 (2020) 185–194, https://doi. peptides: mechanisms of action and synergy with drugs against fungal pathogens,
org/10.1016/j.fsi.2020.04.001. Front. Microbiol. 8 (2017) 2, https://doi.org/10.3389/fmicb.2017.00002.
[97] Z. Meng, B. Zhang, B. Liu, H. Li, S. Fan, D. Yu, High carotenoids content can [120] V. Morshedi, B. Bojarski, S. Hamedi, H. Torahi, G. Hashemi, C. Faggio, Effects of
enhance resistance of selected Pinctada fucata families to high temperature stress, dietary bovine lactoferrin on growth performance and immuno-physiological
Fish Shellfish Immunol. 61 (2017) 211–218, https://doi.org/10.1016/j. responses of Asian sea bass (Lates calcarifer) fingerlings, Probiot. Antimicrob.
fsi.2016.12.032. Protein 13 (6) (2021) 1790–1797. https://link.springer.com/article/10.1007/s12
602-021-09805-4.

305
S. Vijayaram et al. Fish and Shellfish Immunology 130 (2022) 294–308

[121] H.L. Xia, J. Cheng, D.P. Yu, W.J. Chen, Y.S. Lu, Research progress on [141] S. Yilmaz, E. Yilmaz, M.A. Dawood, E. Ringø, E. Ahmadifar, H.M. Abdel-Latif,
peptidoglycan recognition proteins in fish, Biotechnol. Bull. 34 (8) (2018) 58, Probiotics, prebiotics, and synbiotics used to control vibriosis in fish: a review,
https://doi.org/10.13560/j.cnki.biotech.bull.1985.2018-0218. Aquaculture 547 (2022), 737514, https://doi.org/10.1016/j.
[122] S. Roy, P. Bossier, P. Norouzitallab, D. Vanrompay, Trained immunity and Aquaculture..2021.737514.
perspectives for shrimp aquaculture, Rev. Aquacult. 12 (4) (2020) 2351–2370, [142] K. Mohan, T. Muralisankar, V. Uthayakumar, R. Chandirasekar, N. Revathi, A.
https://doi.org/10.1111/raq.12438. R. Ganesan, P. Seedevi, Trends in the extraction, purification, characterisation
[123] H.L. Yang, X. Hu, J.D. Ye, V. Seerengaraj, W. Yang, C.X. Ai, Y.Z. Sun, Cell wall and biological activities of polysaccharides from tropical and sub-tropical fruits–A
components of Bacillus pumilus SE5 improved the growth, digestive and immunity comprehensive review, Carbohydr. Polym. 238 (2020), 116185, https://doi.org/
of grouper (Epinephelus coioides), Curr. Chin. Sci. 1 (2) (2021) 231–239. https 10.1016/j.carbpol.2020.116185.
://www.eurekaselect.com/node/189018/article/cell-wall-components-of-bacill [143] F. Motta, M.E. Gershwin, C. Selmi, Mushrooms and immunity, J. Autoimmun. 117
us-pumilus-se5-improved-the-growth-digestive-and-immunity-of-grouper-epi (2021), 102576, https://doi.org/10.1016/j.jaut.2020.102576.
nephelus-coioides. [144] B. Deng, Z.P. Wang, W.J. Tao, W.F. Li, C. Wang, M.Q. Wang, D. Wu, Effects of
[124] H. Kondo, T. Kikumoto, K. Yoshii, N. Murase, H. Yamada, Y. Fukuda, I. Hirono, polysaccharides from mycelia of Cordyceps sinensis on growth performance,
Effects of peptidoglycan and polyinosinic: polycytidylic acid on the recombinant immunity and antioxidant indicators of the white shrimp Litopenaeus vannamei,
subunit vaccine efficacy against Edwardsiella tarda in Japanese flounder Aquacult. Nutr. 21 (2) (2015) 173–179, https://doi.org/10.1111/anu.12147.
Paralichthys olivaceus, Fish Pathol. 56 (3) (2021) 149–155, https://doi.org/ [145] K. Mohan, A.M. Padmanaban, V. Uthayakumar, R. Chandirasekar,
10.3147/jsfp.56.149. T. Muralisankar, P. Santhanam, Effect of dietary Ganoderma lucidum
[125] C.Y. Ang, M. Sano, S. Dan, M. Leelakriangsak, T.M. Lal, Postbiotics applications as polysaccharides on biological and physiological responses of the giant freshwater
infectious disease control agent in aquaculture, Biocontrol Sci. 25 (1) (2020) 1–7, prawn Macrobrachium rosenbergii, Aquaculture 464 (2016) 42–49, https://doi.
https://doi.org/10.4265/bio.25.1. org/10.1016/j.Aquaculture..2016.05.046.
[126] Z. Hu, X. Cao, M. Guo, C. Li, Identification and characterization of a novel short- [146] G. Mohammadi, M. Hafezieh, A.A. Karimi, M.N. Azra, H. Van Doan,
type peptidoglycan recognition protein in Apostichopus japonicus, Fish Shellfish W. Tapingkae, M.A. Dawood, The synergistic effects of plant polysaccharide and
Immunol. 99 (2020) 257–266, https://doi.org/10.1016/j.fsi.2020.02.013. Pediococcus acidilactici as a synbiotic additive on growth, antioxidant status,
[127] C.R. de Cruz, F.Y. Yamamoto, M. Ju, K. Chen, A. Velasquez, D.M. Gatlin III, immune response, and resistance of Nile tilapia (Oreochromis niloticus) against
Efficacy of purified nucleotide supplements on the growth performance and Aeromonas hydrophila, Fish Shellfish Immunol. 120 (2022) 304–313, https://doi.
immunity of hybrid striped bass Morone chrysops x Morone saxatilis, Fish org/10.1016/j.fsi.2021.11.028.
Shellfish Immunol. 98 (2020) 868–874, https://doi.org/10.1016/j. [147] H. Do Huu, H.M. Sang, N.T.T. Thuy, Dietary β-glucan improved growth
fsi.2019.11.046. performance, Vibrio counts, haematological parameters and stress resistance of
[128] A.P.D.C. Alves, R.R. Paulino, R.T. Pereira, D.V. da Costa, P.V. e Rosa, Nile tilapia pompano fish, Trachinotus ovatus Linnaeus, 1758, Fish Shellfish Immunol. 54
fed insect meal: growth and innate immune response in different times under (2016) 402–410, https://doi.org/10.1016/j.fsi.2016.03.161.
lipopolysaccharide challenge, Aquacult. Res. 52 (2) (2021) 529–540, https://doi. [148] X. Xu, X. Zhang, Lentinula edodes-derived polysaccharide alters the spatial
org/10.1111/are.14911. structure of gut microbiota in mice, PLoS One 10 (1) (2015), e0115037, https://
[129] S.W. Luo, N.X. Xiong, Z.Y. Luo, K.K. Luo, S.J. Liu, C. Wu, M. Wen, Effect of doi.org/10.1371/journal.pone.0115037.
Lipopolysaccharide (LPS) stimulation on apoptotic process and oxidative stress in [149] G. Ren, M. Yu, K. Li, Y. Hu, Y. Wang, X. Xu, J. Qu, Seleno-lentinan prevents
fibroblast cell of hybrid crucian carp compared with those of Carassius cuvieri chronic pancreatitis development and modulates gut microbiota in mice, J. Funct.
and Carassius auratus red var, Comp. Biochem. Physiol. C Toxicol. Pharmacol. Foods 22 (2016) 177–188, https://doi.org/10.1016/j.jff.2016.01.035.
248 (2021), 109085, https://doi.org/10.1016/j.cbpc.2021.109085. [150] S. Shahbazi, A. Bolhassani, Immunostimulants: types and functions, J. Med.
[130] M.F. Abd El-Kader, M. Shukry, M.A. Dawood, M.M. Hassan, F. Farrag, A. Omar, E. Microbiol. Infect. Dis. 4 (3) (2016) 45–51. https://jommid.pasteur.ac.ir/ar
M. Moustafa, Ameliorative effect of dietary lipopolysaccharides on Oreochromis ticle-1-131-en.html.
niloticus juveniles submitted to aflatoxin B1-induced oxidative stress and bacterial [151] S. Xiang, X. Chen, J. Wang, H. Peng, X. Lv, L. Jiang, L. Ma, A lentinan-loaded
challenge, Aquacult. Res. 52 (8) (2021) 3660–3676, https://doi.org/10.1111/ hydrogel with core-shell structure induces broad-spectrum resistance against
are.15211. plant virus by activating the expression of CML30, Authorea Preprints, https
[131] E. Holen, M.H. Austgulen, M. Espe, RNA form baker’s yeast cultured with and ://www.authorea.com/doi/full/10.22541/au.160503099.94865844, 2020.
without lipopolysaccharide (LPS) modulates gene transcription in an intestinal [152] W. Yao, X. Li, C. Zhang, J. Wang, Y. Cai, X. Leng, Effects of dietary synbiotics
epithelial cell model, RTgutGC from rainbow trout (Oncorhynchus mykiss), Fish supplementation methods on growth, intestinal health, non-specific immunity
Shellfish Immunol. 119 (2021) 397–408, https://doi.org/10.1016/j. and disease resistance of Pacific white shrimp, Litopenaeus vannamei, Fish
fsi.2021.10.018. Shellfish Immunol. 112 (2021) 46–55, https://doi.org/10.1016/j.
[132] G. Mohammadi, T.J. Adorian, G. Rafiee, Beneficial effects of Bacillus subtilis on fsi.2021.02.011.
water quality, growth, immune responses, endotoxemia and protection against [153] F. Li, F. Gao, J. Tan, C. Fan, H. Sun, J. Yan, X. Wang, Characterization and
lipopolysaccharide-induced damages in Oreochromis niloticus under biofloc identification of enzyme-producing microflora isolated from the gut of sea
technology system, Aquacult. Nutr. 26 (5) (2020) 1476–1492, https://doi.org/ cucumber Apostichopus japonicus, Chin. J. Oceanol. Limnol. 34 (1) (2016)
10.1111/anu.13096. 153–162, https://doi.org/10.1007/s00343-015-4149-z.
[133] J. Xue, K. Shen, Y. Hu, Y. Hu, V. Kumar, G. Yang, C. Wen, Effects of dietary [154] R. Yuji-Sado, F. Raulino-Domanski, P.F. de Freitas, F. Baioco-Sales, Growth,
Bacillus cereus, B. subtilis, Paracoccus marcusii, and Lactobacillus plantarum immune status and intestinal morphology of Nile tilapia fed dietary prebiotics
supplementation on the growth, immune response, antioxidant capacity, and (mannan oligosaccharides-MOS), Lat. am. j. aquat. res. 43 (5) (2015) 944–952.
intestinal health of juvenile grass carp (Ctenopharyngodon idellus), Aquacult. Rep. https://www.redalyc.org/pdf/1750/175042668014.pdf.
17 (2020), 100387, https://doi.org/10.1016/j.aqrep.2020.100387. [155] Z.Y. Lu, W.D. Jiang, P. Wu, Y. Liu, S.Y. Kuang, L. Tang, L. Feng, Mannan
[134] H.L. Yang, Y.Z. Sun, X. Hu, J.D. Ye, K.L. Lu, L.H. Hu, J.J. Zhang, Bacillus pumilus oligosaccharides supplementation enhanced head-kidney and spleen immune
SE5 originated PG and LTA tuned the intestinal TLRs/MyD88 signaling and function in on-growing grass carp (Ctenopharyngodon idella), Fish Shellfish
microbiota in grouper (Epinephelus coioides), Fish Shellfish Immunol. 88 (2019) Immunol. 106 (2020) 596–608, https://doi.org/10.1016/j.fsi.2020.08.035.
266–271, https://doi.org/10.1016/j.fsi.2019.03.005. [156] M. Abdel Gayed, H. Elabd, M. Shehabeldin, A. Shaheen, A. El-Asely, A. El-Gawad,
[135] Y. Yang, J. Lim, C. Li, S. Lee, S. Hong, Effects of sulfated polysaccharides isolated A. Abbass, A Review of some prebiotics and probiotics supplementation effects on
from Codium fragile on inflammatory cytokine gene expression and Edwardsiella farmed fishes: with special reference to Mannan oligosaccharides (MOS), Benha
tarda infection in rockfish, Sebastes schlegelii, Fish Shellfish Immunol. 112 (2021) Vet. Med. J. 40 (1) (2021) 141–145, https://doi.org/10.21608/
125–134, https://doi.org/10.1016/j.fsi.2021.03.001. bvmj.2021.62545.1342.
[136] C. Jiang, M. Xu, X. Kuang, J. Xiao, M. Tan, Y. Xie, Y. Wu, Treponema pallidum [157] M.A. Dawood, S. Koshio, S.E. Fadl, H.A. Ahmed, A. El Asely, M.M. Abdel-Daim,
flagellins stimulate MMP-9 and MMP-13 expression via TLR5 and MAPK/NF-κB S. Alkahtani, The modulatory effect of mannanoligosaccharide on oxidative
signaling pathways in human epidermal keratinocytes, Exp. Cell Res. 361 (1) status, selected immune parameters and tolerance against low salinity stress in
(2017) 46–55, https://doi.org/10.1016/j.yexcr.2017.09.040. red sea bream (Pagrus major), Aquacult. Rep. 16 (2020), 100278, https://doi.org/
[137] Q. Gao, Y. Xiao, C. Zhang, M. Min, S. Peng, Z. Shi, Molecular characterization and 10.1016/j.aqrep.2020.100278.
expression analysis of toll-like receptor 2 in response to bacteria in silvery [158] M.N. Akter, K. Zahan, M.A. Zafar, N. Khatun, M.S. Rana, M.I. Mursalin, Effects of
pomfret intestinal epithelial cells, Fish Shellfish Immunol. 58 (2016) 1–9, https:// dietary mannan oligosaccharide on growth performance, feed utilization, body
doi.org/10.1016/j.fsi.2016.08.057. composition and haematological parameters in asian catfish (Clarias batrachus)
[138] Q. Gao, Y. Yue, M. Min, S. Peng, Z. Shi, W. Sheng, T. Zhang, Characterization of juveniles, Turk. J. Fish. Aquat. Sci. 21 (11) (2021) 559–567, https://doi.org/
TLR5 and TLR9 from silver pomfret (Pampus argenteus) and expression profiling in 10.4194/1303-2712-v21_11_04.
response to bacterial components, Fish Shellfish Immunol. 80 (2018) 241–249, [159] S. Ayiku, J. Shen, B.P. Tan, X.H. Dong, H.Y. Liu, Effects of reducing dietary
https://doi.org/10.1016/j.fsi.2018.06.014. fishmeal with yeast supplementations on Litopenaeus vannamei growth, immune
[139] Á. González-Garcinuño, A. Tabernero, Á. Domínguez, M.A. Galán, E.M. Martin del response and disease resistance against Vibrio harveyi, Microbiol. Res. 239 (2020),
Valle, Levan and levansucrases: polymer, enzyme, micro-organisms and 126554, https://doi.org/10.1016/j.micres.2020.126554.
biomedical applications, Biocatal. Biotransform. 36 (3) (2018) 233–244, https:// [160] E. De Marco Castro, P.C. Calder, H.M. Roche, β-1, 3/1, 6-glucans and immunity:
doi.org/10.1080/10242422.2017.1314467. state of the art and future directions, Mol. Nutr. Food Res. 65 (1) (2021),
[140] M.Y. Huang, C.I. Chang, C.C. Chang, L.W. Tseng, C.L. Pan, Effects of dietary levan 1901071, https://doi.org/10.1002/mnfr.201901071.
on growth performance, nonspecific immunity, pathogen resistance and body [161] M.H. Khanjani, G. Ghaedi, M. Sharifinia, Effects of diets containing β-glucan on
composition of orange-spotted grouper (Epinephelus coioides H.), Aquacult. Res. 46 survival, growth performance, haematological, immunity and biochemical
(11) (2015) 2752–2767. https://onlinelibrary.wiley.com/doi/abs/10.1111/are. parameters of rainbow trout (Oncorhynchus mykiss) fingerlings, Aquacult. Res. 53
12430. (5) (2022) 1842–1850, https://doi.org/10.1111/are.15712.

306
S. Vijayaram et al. Fish and Shellfish Immunology 130 (2022) 294–308

[162] A. Chhetri, The role of (1, 3)-β-d-Glucan synthase, in: Fungal Cell Wall Biosynthesis [181] G. Shanmugavel, G. Krishnamoorthy, Mucuna pruriens-seed extract stimulating
(Doctoral Dissertation, Duke University, 2020. https://www.proquest.com/ope immune system in Litopenaeus vannamei against Vibrio harveyi, World News Nat.
nview/7ad737cf781564ac5c8d4b24e57de6f6/1?pq-origsite=gscholar&cbl Sci. 39 (2021) 95–112. https://bibliotekanauki.pl/articles/1839790.
=18750&diss=y. [183] D.S. Lakshmi, S. Sankaranarayanan, T.K. Gajaria, G. Li, W. Kujawski, J. Kujawa,
[163] R. Pogue, E.J. Murphy, G.W. Fehrenbach, E. Rezoagli, N.J. Rowan, Exploiting R. Navia, A short review on the valorization of green seaweeds and ulvan:
immunomodulatory properties of β-glucans derived from natural products for feedstock for chemicals and biomaterials, Biomolecules 10 (7) (2020) 991,
improving health and sustainability in aquaculture-farmed organisms: concise https://doi.org/10.3390/biom10070991.
review of existing knowledge, innovation and future opportunities, Curr. Opin. [184] P. del Rocío Quezada-Rodríguez, E.J. Fajer-Ávila, The dietary effect of ulvan from
Environ. Sci. Health 21 (2021), 100248, https://doi.org/10.1016/j. Ulva clathrata on hematological-immunological parameters and growth of tilapia
coesh.2021.100248. (Oreochromis niloticus), J. Appl. Phycol. 29 (1) (2017) 423–431, https://doi.org/
[164] M.A. Dawood, S. Koshio, M. Ishikawa, S. Yokoyama, Interaction effects of dietary 10.1007/s10811-016-0903-7.
supplementation of heat-killed Lactobacillus plantarum and β-glucan on growth [185] M. Ponce, E. Zuasti, V. Anguís, C. Fernández-Díaz, Effects of the sulfated
performance, digestibility and immune response of juvenile red sea bream, Pagrus polysaccharide ulvan from Ulva ohnoi on the modulation of the immune response
major, Fish Shellfish Immunol. 45 (1) (2015) 33–42, https://doi.org/10.1016/j. in Senegalese sole (Solea senegalensis), Fish Shellfish Immunol. 100 (2020) 27–40,
fsi.2015.01.033. https://doi.org/10.1016/j.fsi.2020.02.054.
[165] M. Marcos-López, H.D. Rodger, Amoebic gill disease and host response in Atlantic [186] R. Harikrishnan, G. Devi, H. Van Doan, C. Balasundaram, J. Arockiaraj,
salmon (Salmo salar L.): a review, Parasite Immunol. 42 (8) (2020), e12766, C. Jagruthi, Efficacy of ulvan on immune response and immuno-antioxidant gene
https://doi.org/10.1111/pim.12766. modulation in Labeo rohita against columnaris disease, Fish Shellfish Immunol.
[166] J.Y. Loh, K.S. Lai, P.T. Lee, H.J. Liew, A.S.Y. Ting, Effects of Artemia nauplii 117 (2021) 262–273, https://doi.org/10.1016/j.fsi.2021.08.004.
bioencapsulated with Lactococcus lactis subsp. lactis CF4MRS and sodium [187] Q.D. Lauzon, A.E. Serrano Jr., Ulvan extract from Enteromorpha intestinalis
alginate on edwardsiellosis protection and digestive enzyme production in enhances immune responses in Litopenaeus vannamei and Penaeus monodon
climbing perch larvae, Anabas testudineus (Bloch, 1792), J. Appl. Aquacult. juveniles, Anim. Biol. Anim. Husb. 7 (1) (2015) 1–10. https://d1wqtxts1xzle7.
(2021) 1–18, https://doi.org/10.1080/10454438.2021.1935389. cloudfront.net/37271090/.
[167] J.Y. Loh, H.K. Chan, H.C. Yam, L.L.A. In, C.S.Y. Lim, An overview of the [188] M. He, G. Liu, Y. Liu, K. Yang, X. Qi, A. Huang, E. Wang, Effects of geniposide as
immunomodulatory effects exerted by probiotics and prebiotics in grouper fish, immunostimulant on the innate immune response and disease resistance in
Aquacult. Int. 28 (2) (2020) 729–750. https://link.springer.com/article/10.1007 crucian carp, Aquaculture 529 (2020), 735713, https://doi.org/10.1016/j.
/s10499-019-00491-2. aquaculture.2020.735713.
[168] M. Chen, X.Q. Chen, L.X. Tian, Y.J. Liu, J. Niu, Beneficial impacts on growth, [189] C. Qin, Z. Zhang, Y. Wang, S. Li, C. Ran, J. Hu, Z. Zhou, EPSP of L. Casei BL23
intestinal health, immune responses and ammonia resistance of pacific white protected against the infection caused by Aeromonas veronii via enhancement of
shrimp (Litopenaeus vannamei) fed dietary synbiotic (mannan oligosaccharide and immune response in zebrafish, Front. Microbiol. 8 (2017) 2406. https://www.
Bacillus licheniformis), Aquacult. Rep. 17 (2020), 100408, https://doi.org/ frontiersin.org/articles/10.3389/fmicb.2017.02406/full.
10.1016/j.aqrep.2020.100408. [190] C.Y. Wang, Z.B. Li, Growth performance, digestive enzyme activity and immune
[169] V.T. Ojerio, V.L. Corre, N.A. Toledo, K.G.S. Andrino-Felarca, L.M. Nievales, R.F. response of Japanese sea bass, Lateolabrax japonicus fed with
M. Traifalgar, Alginic acid as immunostimulant: effects of dose and frequency on fructooligosaccharide, Aquacult. Nutr. 26 (2) (2020) 296–305, https://doi.org/
growth performance, immune responses, and white spot syndrome virus 10.1111/anu.12990.
resistance in tiger shrimp Penaeus monodon (Fabricius, 1798), Aquacult. Int. 26 [191] A. de Lima Paz, J.M. da Silva, K.M.M. da Silva, A.L. Val, Protective effects of the
(1) (2018) 267–278. https://link.springer.com/article/10.1007/s10499-017-0 fructooligosaccharide on the growth performance, hematology, immunology
212-4. indicators and survival of tambaqui (Colossoma macropomum, Characiformes:
[170] H. Tayefi Nasrabadi, M. Heidarieh, N. Sheikhzadeh, S. Niksefat Zendeh, P. Karimi, serrasalmidae) infected by Aeromonas hydrophila, Aquacult. Rep. 15 (2019),
Effects of irradiated Ergosan on growth performance, serum biochemical 100222, https://doi.org/10.1016/j.aqrep.2019.100222.
parameters and skin mucosal immune responses in crucian carp (Carassius [192] L. Hu, H. Yang, Y. Zhao, J. Ye, K. Lu, Y. Sun, Effects of fructooligosaccharide on
carassius), Aquat. Physiol. Biotechnol. 9 (2) (2021) 107–124, https://doi.org/ immune function and intestinal morphology of grouper Epinephelus coioides,
10.22124/japb.2021.17238.1389. Fish. Sci. 38 (5) (2019) 589–594. https://www.cabdirect.org/cabdirect/abstract/
[171] F.S. Farooqi, W.U.H. Qureshi, Immunostimulants for Aquaculture. health 20193471302.
management, J. Pharmacogn. Phytochem. 7 (2018) 1441–1447. https://www.ph [193] E. Pietrzak, J. Mazurkiewicz, A. Slawinska, Innate immune responses of skin
ytojournal.com/archives/2018/vol7issue6/PartY/7-5-684-797.pdf. mucosa in common carp (Cyprinus carpio) fed a diet supplemented with
[172] E. Vallejos-Vidal, F. Reyes-López, M. Teles, S. MacKenzie, The response of fish to galactooligosaccharides, Animals 10 (3) (2020) 438. https://www.mdpi.com
immunostimulant diets, Fish Shellfish Immunol. 56 (2016) 34–69, https://doi. /2076-2615/10/3/438.
org/10.1016/j.fsi.2016.06.028. [194] A. Mustafa, A. Buentello, D. Gatlin, D. Lightner, M. Hume, A. Lawrence, Dietary
[173] D.L. Prabu, N.P. Sahu, A.K. Pal, S. Dasgupta, A. Narendra, Immunomodulation supplementation of galactooligosaccharides (GOS) in Pacific white shrimp,
and interferon gamma gene expression in sutchi cat fish, Pangasianodon Litopenaeus vannamei, cultured in a recirculating system and its effects on gut
hypophthalmus: effect of dietary fucoidan rich seaweed extract (FRSE) on pre and microflora, growth, stress, and immune response, J. Immunoassay Immunochem.
post challenge period, Aquacult. Res. 47 (1) (2016) 199–218, https://doi.org/ 40 (6) (2019) 662–675, https://doi.org/10.1080/15321819.2019.1675694.
10.1111/are.12482. [195] A. Ajdari, H. Ghafarifarsani, S.H. Hoseinifar, S. Javahery, F. Narimanizad,
[174] M. Saeed, M.A. Arain, S. Ali Fazlani, I.B. Marghazani, M. Umar, J. Soomro, K. Gatphayak, H. Van Doan, Effects of dietary supplementation of PrimaLac,
M. Alagawany, A comprehensive review on the health benefits and nutritional inulin, and biomin imbo on growth performance, antioxidant, and innate immune
significance of fucoidan polysaccharide derived from brown seaweeds in human, responses of common carp (Cyprinus carpio), Aquacult. Nutr. (2022), https://doi.
animals and aquatic organisms, Aquacult. Nutr. 27 (3) (2021) 633–654, https:// org/10.1155/2022/8297479, 2022.
doi.org/10.1111/anu.13233. [196] L. Zhou, J. Zhang, M. Yan, S. Tang, X. Wang, J.G. Qin, E. Li, Inulin alleviates
[175] C.H. Hsu, J.C. Chen, Y.C. Lin, Y.Y. Chen, P.C. Liu, B.W. Lin, J.F. Hsieh, White hypersaline-stress induced oxidative stress and dysbiosis of gut microbiota in Nile
shrimp Litopenaeus vannamei that have received mixtures of heat-killed and tilapia (Oreochromis niloticus), Aquaculture 529 (2020), 735681, https://doi.
formalin-inactivated Vibrio alginolyticus and V. harveyi exhibit recall memory and org/10.1016/j.aquaculture.2020.735681.
show increased phagocytosis and resistance to Vibrio infection, Fish Shellfish [197] A.G. Huang, W.H. He, F.L. Zhang, C.S. Wei, Y.H. Wang, Natural component
Immunol. 112 (2021) 151–158, https://doi.org/10.1016/j.fsi.2020.11.013. geniposide enhances survival rate of crayfish Procambarus clarkii infected with
[176] R. Salehpour, N.A. Biuki, M. Mohammadi, A. Dashtiannasab, P. Ebrahimnejad, white spot syndrome virus, Fish Shellfish Immunol. (2022), https://doi.org/
The dietary effect of fucoidan extracted from brown seaweed, Cystoseira trinodis 10.1016/j.fsi.2022.05.037.
(C. Agardh) on growth and disease resistance to WSSV in shrimp Litopenaeus [198] S.H. Hoseinifar, M.A. Jahazi, N. Nikdehghan, H. Van Doan, M.G. Volpe,
vannamei, Fish Shellfish Immunol. 119 (2021) 84–95, https://doi.org/10.1016/j. M. Paolucci, Effects of dietary polyphenols from agricultural by-products on
fsi.2021.09.005. mucosal and humoral immune and antioxidant responses of convict cichlid
[177] S.U. Kadam, C.P. O’Donnell, D.K. Rai, M.B. Hossain, C.M. Burgess, D. Walsh, B. (Amatitlania nigrofasciata), Aquaculture 517 (2020), 734790, https://doi.org/
K. Tiwari, Laminarin from Irish brown seaweeds Ascophyllum nodosum and 10.1016/j.aquaculture.2019.734790.
Laminaria hyperborea: ultrasound assisted extraction, characterization and [199] A. Ahmadi, D. Bagheri, S.H. Hoseinifar, V. Morshedi, M. Paolucci, Beneficial role
bioactivity, Mar. Drugs 13 (7) (2015) 4270–4280, https://doi.org/10.3390/ of polyphenols as feed additives on growth performances, immune response and
md13074270. antioxidant status of Lates Calcarifer (Bloch, 1790) juveniles, Aquaculture 552
[178] S.U. Kadam, B.K. Tiwari, C.P. O’Donnell, Extraction, structure and biofunctional (2022), 737955, https://doi.org/10.1016/j.aquaculture.2022.737955.
activities of laminarin from brown algae, Int. J. Food Sci. Technol. 50 (1) (2015) [200] L. Zhao, B.D. Yuan, J.L. Zhao, N. Jiang, A.Z. Zhang, G.Q. Wang, M.Y. Li,
24–31, https://doi.org/10.1111/ijfs.12692. Amelioration of hexavalent chromium-induced bioaccumulation, oxidative stress,
[179] Y. Meng, F. Lyu, X. Xu, L. Zhang, Recent advances in chain conformation and tight junction proteins and immune-related signaling factors by Allium
bioactivities of triple-helix polysaccharides, Biomacromolecules 21 (5) (2020) mongolicum Regel flavonoids in Ctenopharyngodon idella, Fish Shellfish
1653–1677, https://doi.org/10.1021/acs.biomac.9b01644. Immunol. 106 (2020) 993–1003, https://doi.org/10.1016/j.fsi.2020.09.005.
[180] C. Cui, L. Zhu, X. Tang, J. Xing, X. Sheng, H. Chi, W. Zhan, Differential white spot [201] P. Bussabong, T. Rairat, N. Chuchird, A. Keetanon, P. Phansawat,
syndrome virus-binding proteins in two hemocyte subpopulations of Chinese K. Cherdkeattipol, W. Kraitavin, Effects of isoquinoline alkaloids from Macleaya
shrimp (Fenneropenaeus chinensis), Dev. Comp. Immunol. 125 (2021), 104215, cordata on growth performance, survival, immune response, and resistance to
https://doi.org/10.1016/j.dci.2021.104215. Vibrio parahaemolyticus infection of Pacific white shrimp (Litopenaeus
vannamei), PLoS One 16 (5) (2021), e0251343. https://journals.plos.org/plosone
/article?id=10.1371/journal.pone.0251343.

307
S. Vijayaram et al. Fish and Shellfish Immunology 130 (2022) 294–308

[202] J. Jiang, S. Gao, X. Wang, X. Guan, B. Wang, Z. Chen, Z. Zhou, The role of a novel [206] C. Ma, H. Guo, H. Chang, S. Huang, S. Jiang, D. Huo, X. Zhu, The effects of
secretory peptidoglycan recognition protein from the sea cucumber Apostichopus exopolysaccharides and exopolysaccharide-producing Lactobacillus on the
japonicus in innate immunity, Aquaculture 546 (2022), 737339, https://doi.org/ intestinal microbiome of zebrafish (Danio rerio), BMC Microbiol. 20 (1) (2020)
10.1016/j.aquaculture.2021.737339. 1–11. https://bmcmicrobiol.biomedcentral.com/articles/10.1186/s12866-02
[203] X. Li, S. Yuan, Z. Sun, L. Lei, S. Wan, J. Wang, Q. Gao, Gene identification and 0-01990-6.
functional analysis of peptidoglycan recognition protein from the spotted sea [207] S.K. Gupta, Ameliorative and protective effects of prebiotic, microbial levan in
bass, Fish Shellfish Immunol. 106 (2020) 1014–1024. https://www.sciencedirect. common carp,(Cyprinus carpio) fry under experimental exposure to fipronil, Int.
com/science/article/abs/pii/S1050464820306057. J. Aquat. Biol. 9 (2) (2021) 134–147. https://ij-aquaticbiology.com/index.
[204] M.Y. Li, X.M. Zhu, X.T. Niu, X.M. Chen, J.X. Tian, Y.D. Kong, G.Q. Wang, Effects php/ijab/article/view/866.
of dietary Allium mongolicum Regel polysaccharide on growth, [208] S. Anusha, T. Neeraja, P. Haribabu, P. Akshaya, Effect of different biofloc based
lipopolysaccharide-induced antioxidant responses and immune responses in culture systems on the growth and immune response of Tilapia (Oreochromis
Channa argus, Mol. Biol. Rep. 46 (2) (2019) 2221–2230. https://link.springer. niloticus), Survival 2 (2020) 63. https://www.fisheriesjournal.com/archiv
com/article/10.1007/s11033-019-04677-y. es/2020/vol8issue6/PartC/8-5-67-508.pdf.
[205] Y. Risjani, N. Mutmainnah, P. Manurung, S.N. Wulan, Exopolysaccharide from [209] C. Carballo, P.I. Pinto, A.P. Mateus, C. Berbel, C.C. Guerreiro, J.F. Martinez-
Porphyridium cruentum (purpureum) is not toxic and stimulates immune Blanch, M. Manchado, Yeast β-glucans and microalgal extracts modulate the
response against vibriosis: the assessment using zebrafish and white shrimp immune response and gut microbiome in Senegalese sole (Solea senegalensis),
Litopenaeus vannamei, Mar. Drugs 19 (3) (2021) 133. https://www.mdpi.com Fish Shellfish Immunol. 92 (2019) 31–39, https://doi.org/10.1016/j.
/1660-3397/19/3/133. fsi.2019.05.044.

308

You might also like